1
|
Takamiya A, Vande Casteele T, Bouckaert F, Van Cauwenberge MG, Laroy M, De Winter FL, Dupont P, Van den Stock J, Koole M, Van Laere K, Emsell L, Vandenbulcke M. Accelerated aging of white matter in late-life depression: evidence from 18F-flutemetamol PET imaging. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2025:S2451-9022(25)00126-0. [PMID: 40204237 DOI: 10.1016/j.bpsc.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Late-life depression (LLD) is associated with white matter (WM) alterations. Current evidence indicates amyloid PET tracers as sensitive and reliable markers for evaluating normal-appearing WM (NAWM) on magnetic resonance imaging (MRI), showing an association between lower uptake and Alzheimer's disease pathology and higher uptake with age-related changes. Utilizing this novel and reliable technique, we aimed to distinguish two hypothetical models for neurobiology of LLD: the pathological neurodegenerative model and the accelerated aging model. METHODS In this monocentric cross-sectional study, a total of 103 participants, including 61 patients with LLD (age 73.8±7.0 years, 41 female) and 42 healthy controls (age 72.5±7.6 years, 28 female), underwent PET imaging with 18F-flutemetamol, MRI, and clinical assessment. T2-weighted fluid-attenuated inversion recovery (FLAIR) images were segmented into WM hyperintensities (WMH) and NAWM. RESULTS 18F-flutemetamol standardized uptake value ratio (SUVR) in WMH was significantly lower than that in NAWM (t=7.8, df=102, p<0.001). Compared to healthy controls, patients with LLD exhibited higher 18F-flutemetamol SUVR in both NAWM (p<0.001, Cohen's d=0.91) and WMH (p=0.005, d=0.56), even after controlling for age and 18F-flutemetamol SUVR in cortical gray matter. CONCLUSIONS Our result of elevated 18F-flutemetamol uptake in NAWM does not align with the pathological neurodegenerative aging pattern observed in Alzheimer's disease but is in line with patterns of age-related changes. This distinction is crucial for the development of future targeted treatments.
Collapse
Affiliation(s)
- Akihiro Takamiya
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; Hills Joint Research Laboratory for Future Preventive Medicine and Wellness, Keio University School of Medicine, Tokyo, Japan.
| | - Thomas Vande Casteele
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium
| | - Filip Bouckaert
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; University Psychiatric Center KU Leuven, Department of Geriatric Psychiatry, Leuven, Belgium
| | | | - Maarten Laroy
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium
| | - François-Laurent De Winter
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; University Psychiatric Center KU Leuven, Department of Geriatric Psychiatry, Leuven, Belgium
| | - Patrick Dupont
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; KU Leuven, Leuven Brain Institute, Department of Neurosciences, Laboratory for Cognitive Neurology, Leuven, Belgium
| | - Jan Van den Stock
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; University Psychiatric Center KU Leuven, Department of Geriatric Psychiatry, Leuven, Belgium
| | - Michel Koole
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Nuclear Medicine, Leuven, Belgium
| | - Koen Van Laere
- KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Nuclear Medicine, Leuven, Belgium; Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Louise Emsell
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; University Psychiatric Center KU Leuven, Department of Geriatric Psychiatry, Leuven, Belgium; KU Leuven, Leuven Brain Institute, Department of Imaging and Pathology, Translational MRI, Leuven, Belgium
| | - Mathieu Vandenbulcke
- KU Leuven, Leuven Brain Institute, Department of Neurosciences, Neuropsychiatry, Leuven, Belgium; University Psychiatric Center KU Leuven, Department of Geriatric Psychiatry, Leuven, Belgium
| |
Collapse
|
2
|
Kamal F, Moqadam R, Morrison C, Dadar M. Racial and ethnic differences in white matter hypointensities: The role of vascular risk factors. Alzheimers Dement 2025; 21:e70105. [PMID: 40145319 PMCID: PMC11947760 DOI: 10.1002/alz.70105] [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: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION White matter hypointensities (WMHs) are markers of cerebrovascular pathology associated with cognitive decline. Reports of racial and ethnic differences in WMHs have been inconsistent across studies. This study examined whether race and ethnicity influence WMH burden and whether vascular risk factors explain these differences. METHODS Data from the National Alzheimer's Coordinating Center included 7132 Whites, 892 Blacks, 283 Asians, and 661 Hispanics. Baseline and longitudinal WMHs were examined using linear regression and mixed-effects models across racial and ethnic groups, controlling for demographics and vascular risk factors. RESULTS Adjusting for vascular risk factors reduced WMH burden differences and eliminated differences in temporal regions in Black versus White older adults. For Hispanics, differences became significant after adjusting for vascular risk factors. DISCUSSION Although some racial and ethnic WMH disparities are influenced by vascular risk factors, others persist, highlighting the need for multidimensional approaches when targeting WMHs in diverse populations. HIGHLIGHTS Current research is inconsistent as to whether there are racial differences in white matter hypointensities (WMHs). Blacks exhibit higher WMH burden than Whites, mediated by vascular factors. In Hispanics, WMH differences emerged only after adjusting for vascular risk factors.
Collapse
Affiliation(s)
- Farooq Kamal
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteVerdunQuebecCanada
| | - Roqaie Moqadam
- Douglas Mental Health University InstituteVerdunQuebecCanada
| | | | - Mahsa Dadar
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteVerdunQuebecCanada
| |
Collapse
|
3
|
Kasahara H, Ikeda M, Makioka K, Nakamura T, Takahashi R, Kawarabayashi T, Sanada E, Suto T, Higuchi T, Tsushima Y, Ikeda Y. White matter lesions rather than amyloid-β burden are associated with cognitive decline in Alzheimer's disease. J Alzheimers Dis 2025; 104:319-324. [PMID: 39956952 DOI: 10.1177/13872877251316516] [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] [Indexed: 02/18/2025]
Abstract
We assessed the severity of amyloid-β accumulation and white matter lesions (WML) in the brain, and their association with cognitive decline, in patients (n = 47) with Alzheimer's disease (AD). The mean cortical standardized uptake value ratio (mcSUVR) was derived from amyloid positron emission tomography, while the percentage of WML area relative to brain parenchymal area was determined using magnetic resonance imaging. Cognitive decline was positively correlated with WML severity (r = 0.50, p = 0.042) but not mcSUVR (Aβ accumulation). Managing vascular risk factors can prevent cognitive decline in patients with AD.
Collapse
Affiliation(s)
- Hiroo Kasahara
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masaki Ikeda
- Division of General Education (Neurology), Faculty of Health and Medical Care, Saitama Medical University, Hidaka, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takumi Nakamura
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoma Takahashi
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takeshi Kawarabayashi
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Etsuko Sanada
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takayuki Suto
- Department of Radiology, Gunma University Hospital, Maebashi, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
4
|
Miner AE, Groh JR, Farris C, Hattiangadi S, Cui A, Brickman AM, Alshikho M, Rabinovici GD, Rosen HJ, Cobigo Y, Asken B, Nowinski CJ, Bureau S, Shahrokhi F, Tripodis Y, Ly M, Altaras C, Lenio S, Stern RA, Rosen G, Kelley H, Huber BR, Stein TD, Mez J, McKee AC, Alosco ML. Does white matter and vascular injury from repetitive head impacts lead to a novel pattern on T2 FLAIR MRI? A hypothesis proposal and call for research. Alzheimers Dement 2025; 21:e70085. [PMID: 40145364 PMCID: PMC11947747 DOI: 10.1002/alz.70085] [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: 09/12/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/28/2025]
Abstract
The goal of this paper is to introduce the hypothesis that white matter (WM) and vascular injury are long-term consequences of repetitive head impacts (RHI) that result in a novel T2 fluid attenuated inversion recovery (FLAIR) magnetic resonance imaging pattern. A non-systematic literature review of autopsy and FLAIR studies of RHI-exposed adults was first conducted as a foundation for our hypothesis. A case series of RHI-exposed participants is presented to illustrate the unique FLAIR WM hyperintensities (WMH) pattern. Current literature shows a direct link between RHI and later-life WM/vascular neuropathologies, and that FLAIR WMH are associated with RHI, independent of modifiable vascular risk factors. Initial observations suggest a distinctive pattern of WMH in RHI-exposed participants, termed RHI-associated WMH (RHI-WMH). RHI-WMH defining features are as follows: (1) small, punctate, non-confluent, (2) spherical, and (3) proximal to the gray matter. Our hypothesis serves as a call for research to empirically validate RHI-WMH and clarify their biological and clinical correlates. HIGHLIGHTS: Repetitive head impacts (RHI) have been associated with later-life white matter (WM) and vascular neuropathologies. T2 FLAIR MRI of RHI-exposed participants reveals a potentially unique WM hyperintensity (WMH) pattern that is termed RHI-associated WMH (RHI-WMH). RHI-WMH are characterized as (1) small, punctate, and non-confluent, (2) spherical, and (3) proximal to the gray matter at an area anatomically susceptible to impact injury, such as the depths of the cortical sulci.
Collapse
|
5
|
DeCarli C, Rajan KB, Jin LW, Hinman J, Johnson DK, Harvey D, Fornage M. WMH Contributions to Cognitive Impairment: Rationale and Design of the Diverse VCID Study. Stroke 2025; 56:758-776. [PMID: 39545328 PMCID: PMC11850211 DOI: 10.1161/strokeaha.124.045903] [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] [Indexed: 11/17/2024]
Abstract
As awareness of dementia increases, more individuals with minor cognitive complaints are requesting clinical assessment. Neuroimaging studies frequently identify incidental white matter hyperintensities, raising patient concerns about their brain health and future risk for dementia. Moreover, current US demographics indicate that ≈50% of these individuals will be from diverse backgrounds by 2060. Racial and ethnic minority populations bear a disproportionate burden of vascular risk factors magnifying dementia risk. Despite established associations between white matter hyperintensities and cognitive impairment, including dementia, no study has comprehensively and prospectively examined the impact of individual and combined magnetic resonance imaging measures of white matter injury, their risk factors, and comorbidities on cognitive performance among a diverse, nondemented, stroke-free population with cognitive complaints over an extended period of observation. The Diverse VCID (Diverse Vascular Cognitive Impairment and Dementia) study is designed to fill this knowledge gap through 3 assessments of clinical, behavioral, and risk factors; neurocognitive and magnetic resonance imaging measures; fluid biomarkers of Alzheimer disease, vascular inflammation, angiogenesis, and endothelial dysfunction; and measures of genetic risk collected prospectively over a minimum of 3 years in a cohort of 2250 individuals evenly distributed among Americans of Black/African, Latino/Hispanic, and non-Hispanic White backgrounds. The goal of this study is to investigate the basic mechanisms of small vessel cerebrovascular injury, emphasizing clinically relevant assessment tools and developing a risk score that will accurately identify at-risk individuals for possible treatment or clinical therapeutic trials, particularly individuals of diverse backgrounds where vascular risk factors and disease are more prevalent.
Collapse
Affiliation(s)
- Charles DeCarli
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Kumar B. Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago IL
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine University of California Davis California USA
| | - Jason Hinman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - David K. Johnson
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Danielle Harvey
- Department of Public Health Sciences University of California Davis California USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | |
Collapse
|
6
|
Gao F, Zhan Y, Wang Q, Zhang M, Dai L, Shen Y. Pathological angiogenesis was associated with cerebrovascular lesion and neurodegeneration in Alzheimer's disease. Alzheimers Dement 2025; 21:e14521. [PMID: 39777972 PMCID: PMC11848169 DOI: 10.1002/alz.14521] [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: 09/30/2024] [Revised: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION We investigated the specific factors driving abnormal angiogenesis in Alzheimer's disease (AD) and its role in cerebrovascular lesions and neurodegeneration. METHODS We assessed cerebrovascular pathologies, amyloid-beta (Aβ), and tau pathologies in post mortem human brains and detected 12 angiogenic factors in cerebrospinal fluid (CSF) from the China Aging and Neurodegenerative Disease Initiative (CANDI) cohort. RESULTS We observed severe blood-brain barrier damage and elevated levels of the vascular marker CD31 in human AD brains, which had a stronger correlation with tau pathology than Aβ pathology. Consistently, only CSF pTau181 showed positive associations with CSF angiogenesis factors (soluble vascular endothelial growth factor receptor 2 [sVEGFR2], VEGF-C, VEGF-D, placental growth factor [PLGF], Angiopoietin2, and Serpin E1), but not CSF Aβ42/40. Additionally, higher levels of CSF sVEGFR1, soluble Tyrosine-protein kinase receptor 2 [sTIE2], PLGF, and interleukin 8 [IL8], as well as lower levels of CSF urokinase-type plasminogen activator [uPA], were associated with worsen cerebrovascular pathologies and neurodegeneration. DISCUSSION Our findings indicate that tau pathology may play a critical role in pathological angiogenesis, contributing to cerebrovascular lesions and neurodegeneration in AD. HIGHLIGHTS BBB damage and elevated vascular marker CD31 in human AD brains had a stronger correlation with tau pathology than Aβ pathology. CSF pTau181 mediated the effect of CSF Aβ42/40 on CSF sVEGFR1 and sTIE2. Only CSF pTau181 showed positive associations with sVEGFR2, VEGF-C, VEGF-D, PLGF, Angiopoietin2, and Serpin E1. Higher sVEGFR1, sTIE2, PLGF, and IL8, and lower uPA in CSF, were associated with cerebrovascular pathologies and neurodegeneration.
Collapse
Affiliation(s)
- Feng Gao
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yaxi Zhan
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Qiong Wang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Mengguo Zhang
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Linbin Dai
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yong Shen
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
7
|
Hu HY, Li HQ, Gong WK, Huang SY, Fu Y, Hu H, Dong Q, Cheng W, Tan L, Cui M, Yu JT. Microstructural white matter injury contributes to cognitive decline: Besides amyloid and tau. J Prev Alzheimers Dis 2025; 12:100037. [PMID: 39863331 DOI: 10.1016/j.tjpad.2024.100037] [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: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Cognitive decline and the progression to Alzheimer's disease (AD) are traditionally associated with amyloid-beta (Aβ) and tau pathologies. This study aims to evaluate the relationships between microstructural white matter injury, cognitive decline and AD core biomarkers. METHODS We conducted a longitudinal study of 566 participants using peak width of skeletonized mean diffusivity (PSMD) to quantify microstructural white matter injury. The associations of PSMD with changes in cognitive functions, AD pathologies (Aβ, tau, and neurodegeneration), and volumes of AD-signature regions of interest (ROI) or hippocampus were estimated. The associations between PSMD and the incidences of clinical progression were also tested. Covariates included age, sex, education, apolipoprotein E4 status, smoking, and hypertension. RESULTS Higher PSMD was associated with greater cognitive decline (β=-0.012, P < 0.001 for Mini-Mental State Examination score; β<0, P < 0.05 for four cognitive domains) and a higher risk of clinical progression from normal cognition to mild cognitive impairment (MCI) or AD (Hazard ratio=2.11 [1.38-3.23], P < 0.001). These associations persisted independently of amyloid status. PSMD did not predict changes in Aβ or tau levels, but predicted changes in volumes of AD-signature ROI (β=-0.003, P < 0.001) or hippocampus (β=-0.002, P = 0.010). Besides, the whole-brain PSMD could predict cognitive decline better than regional PSMDs. CONCLUSIONS PSMD may be a valuable biomarker for predicting cognitive decline and clinical progression to MCI and AD, providing insights besides traditional Aβ and tau pathways. Further research could elucidate its role in clinical assessments and therapeutic strategies.
Collapse
Affiliation(s)
- He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Hong-Qi Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Wei-Kang Gong
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, PR China.
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, PR China.
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Mei Cui
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| |
Collapse
|
8
|
Sepulveda-Falla D, Villegas Lanau CA, White III C, Serrano GE, Acosta-Uribe J, Mejía-Cupajita B, Villalba-Moreno ND, Lu P, Glatzel M, Kofler JK, Ghetti B, Frosch MP, Restrepo FL, Kosik KS, Beach TG. Comorbidities in early-onset sporadic versus presenilin-1 mutation-associated Alzheimer disease dementia: Evidence for dependency on Alzheimer disease neuropathological changes. J Neuropathol Exp Neurol 2025; 84:104-113. [PMID: 39656832 PMCID: PMC11747142 DOI: 10.1093/jnen/nlae122] [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] [Indexed: 12/14/2024] Open
Abstract
Studying comorbidities in early onset Alzheimer disease (AD) may provide an advantageous perspective on their pathogenesis because aging factors may be largely inoperative for these subjects. We compared AD comorbidities between early-onset sporadic cases and American and Colombian cases with PSEN1 mutations. AD neuropathological changes (ADNC) were very severe in all groups but more severe in the PSEN1 groups. Lewy body disease and cerebral white matter rarefaction were the most common (up to 60%) of AD comorbidities, followed by arteriolosclerosis (up to 37%), and large-vessel atherosclerosis (up to 20%). Differences between the 3 groups included earlier age of onset in the American PSEN1 cases, shorter disease duration in sporadic cases, and more frequent large-vessel atherosclerosis and cerebral amyloid angiopathy in the Colombian PSEN1 cases. Logistic regression models adjusted for age and sex found the presence of a PSEN1 mutation, an apolipoprotein ε4 allele and TDP-43 pathology to predict an earlier age of onset; Hispanic ethnicity and multiracial subjects were predictive of severe CAA. Comorbidities are common in early onset AD and should be considered when planning clinical trials with such subjects. However, they may be at least partially dependent on ADNC and thus potentially addressable by anti-amyloid or and/anti-tau therapies.
Collapse
Affiliation(s)
- Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Charles White III
- Neuropathology Section, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Juliana Acosta-Uribe
- Faculty of Medicine, Neuroscience Group of Antioquia, University of Antioquia, Medellin, Colombia
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, United States
| | - Barbara Mejía-Cupajita
- Faculty of Medicine, Neuroscience Group of Antioquia, University of Antioquia, Medellin, Colombia
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, United States
| | | | - Pinzhang Lu
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | | | - Kenneth S Kosik
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, United States
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, United States
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, United States
| |
Collapse
|
9
|
Malla S, Bryant AG, Jayakumar R, Woost B, Wolf N, Li A, Das S, van Veluw SJ, Bennett RE. Molecular profiling of frontal and occipital subcortical white matter hyperintensities in Alzheimer's disease. Front Neurol 2025; 15:1470441. [PMID: 39845935 PMCID: PMC11753232 DOI: 10.3389/fneur.2024.1470441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/29/2024] [Indexed: 01/24/2025] Open
Abstract
White matter hyperintensities (WMHs) are commonly detected on T2-weighted magnetic resonance imaging (MRI) scans, occurring in both typical aging and Alzheimer's disease (AD). Despite their frequent appearance and their association with cognitive decline in AD, the molecular factors contributing to WMHs remain unclear. In this study, we investigated the transcriptomic profiles of two commonly affected brain regions with coincident AD pathology-frontal subcortical white matter (frontal-WM) and occipital subcortical white matter (occipital-WM)-and compared with age-matched cognitively intact controls. Through RNA-sequencing in frontal- and occipital-WM bulk tissues, we identified an upregulation of genes associated with brain vasculature function in AD white matter. To further elucidate vasculature-specific transcriptomic features, we performed RNA-seq analysis on blood vessels isolated from these white matter regions, which revealed an upregulation of genes related to protein folding pathways. Finally, comparing gene expression profiles between AD individuals with high- versus low-WMH burden showed an increased expression of pathways associated with immune function. Taken together, our study characterizes the diverse molecular profiles of white matter changes in AD and provides mechanistic insights into the processes underlying AD-related WMHs.
Collapse
Affiliation(s)
- Sulochan Malla
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Annie G. Bryant
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- School of Physics, The University of Sydney, Sydney, NSW, Australia
| | - Rojashree Jayakumar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Benjamin Woost
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Nina Wolf
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Andrew Li
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Susanne J. van Veluw
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Rachel E. Bennett
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Kang S, Jeon S, Kim Y, Jeon S, Choi M, Lee Y, Yun M, Ye BS. White matter hyperintensities and cholinergic degeneration as Lewy body disease. Ann Clin Transl Neurol 2025; 12:97-109. [PMID: 39654300 PMCID: PMC11752093 DOI: 10.1002/acn3.52257] [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: 07/23/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Although basal forebrain (BF) cholinergic degeneration and white matter hyperintensities (WMHs) are important in neurodegeneration in Alzheimer's disease (AD) and dementia with Lewy bodies (DLB), their relationships with dopaminergic degeneration and clinical manifestations remain unclear. METHODS A total of 407 patients with cognitive impairment meeting the diagnostic criteria for AD, DLB, or both (AD+DLB) were assessed. All participants underwent 3T MRI, dopamine transporter (DAT) positron emission tomography, neuropsychological tests, and assessments for parkinsonism, cognitive fluctuation, visual hallucination, and rapid eye movement sleep behavior disorder (RBD). General linear and logistic regression models were used to investigate the relationships among BF volume, DAT uptake in the anterior caudate (DAT-AC), WMH volumes in anterior, posterior, periventricular, and deep regions, and clinical manifestations. RESULTS DAT-AC was positively associated with BF volume and negatively associated with anterior periventricular WMH volume, but not with deep WMHs. Both deep and periventricular WMHs volumes were associated with hypertension and the number of microbleeds and lacunae. Lower BF volume and DAT-AC were independently associated with increased risk of cognitive fluctuation and visual hallucination, whereas lower DAT-AC was additionally associated with increased risk of RBD and greater parkinsonian severity. Both lower BF volume and DAT-AC were independently associated with widespread cognitive impairment, whereas higher anterior periventricular WMH volume was associated with executive dysfunction. INTERPRETATION BF cholinergic degeneration and anterior periventricular WMHs are closely associated with dopaminergic degeneration. Anterior periventricular WMHs may represent axonal alterations caused by the interplay between Lewy body-related degeneration and vascular pathologies.
Collapse
Affiliation(s)
- Sungwoo Kang
- Department of NeurologyYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seun Jeon
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Yeoju Kim
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Su‐Hee Jeon
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Minsun Choi
- Metabolism‐Dementia Research InstituteYonsei University College of MedicineSeoul03722Republic of Korea
| | - Young‐gun Lee
- Department of Neurology, Ilsan Paik HospitalInje University College of MedicineGoyang10380Republic of Korea
| | - Mijin Yun
- Department of Nuclear MedicineYonsei University College of MedicineSeoul03722Republic of Korea
| | - Byoung Seok Ye
- Department of NeurologyYonsei University College of MedicineSeoul03722Republic of Korea
| |
Collapse
|
11
|
Silbert LC. Vascular Cognitive Impairment. Continuum (Minneap Minn) 2024; 30:1699-1725. [PMID: 39620840 DOI: 10.1212/con.0000000000001508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OBJECTIVE Vascular cognitive impairment is a major contributor to age-associated cognitive decline, both independently and as a contributor to mixed dementia syndromes. This article reviews the current understanding of how vascular dysfunction contributes to cognitive impairment and dementia risk in older individuals and includes updated diagnostic criteria and treatment recommendations. LATEST DEVELOPMENTS Clinical and research criteria have been evolving to more accurately determine the full prevalence of vascular cognitive impairment. The Boston Criteria version 2.0 for cerebral amyloid angiopathy now includes multiple punctate MRI T2 white matter hyperintensities and MR-visible perivascular spaces in addition to previously described T2* hemorrhagic signatures. MR-visible perivascular spaces are associated with both vascular cognitive impairment and Alzheimer disease, potentially linking cerebrovascular dysfunction to neurodegenerative disorders through its role in brain waste clearance. The American Heart Association's goal for cardiovascular health promotion, "Life's Essential 8," has been updated to include sleep health and acknowledges psychological well-being and social determinants of health as fundamental components necessary to achieve optimal cardiovascular health for all adults. ESSENTIAL POINTS Vascular cognitive impairment is a common and often underrecognized contributor to cognitive impairment in older individuals, with heterogeneous etiologies requiring individualized treatment strategies. Effective cerebrovascular disease risk factor modification starting in midlife is critical to reducing the risk of Alzheimer disease and related dementias, with the goal of preventing vascular brain injury and maintaining cognitive reserve in the presence of nonvascular age-related brain pathologies.
Collapse
|
12
|
Maillard P, Fletcher E, Carmichael O, Schwarz C, Seiler S, DeCarli C. Cerebrovascular markers of WMH and infarcts in ADNI: A historical perspective and future directions. Alzheimers Dement 2024; 20:8953-8968. [PMID: 39535353 DOI: 10.1002/alz.14358] [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: 05/10/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
White matter hyperintensities (WMH) and infarcts found on magnetic resonance imaging (MR infarcts) are common biomarkers of cerebrovascular disease. In this review, we summarize the methods, publications, and conclusions stemming from the Alzheimer's Disease Neuroimaging Initiative (ADNI) related to these measures. We combine analysis of WMH and MR infarct data from across the three main ADNI cohorts with a review of existing literature discussing new methodologies and scientific findings derived from these data. Although ADNI inclusion criteria were designed to minimize vascular risk factors and disease, data across all the ADNI cohorts found consistent trends of increasing WMH volumes associated with advancing age, female sex, and cognitive impairment. ADNI, initially proposed as a study to investigate biomarkers of AD pathology, has also helped elucidate the impact of asymptomatic cerebrovascular brain injury on cognition within a cohort relatively free of vascular disease. Future ADNI work will emphasize additional vascular biomarkers. HIGHLIGHTS: White matter hyperintensities (WMHs) are common to advancing age and likely reflect brain vascular injury among older individuals. WMH and to a lesser extent, magnetic resonance (MR) infarcts, affect risk for transition to cognitive impairment. WMHs and MR infarcts are present, even among Alzheimer's Disease Neuroimaging Initiative (ADNI) participants highly selected to have Alzheimer's disease (AD) as the primary pathology. WMH burden in ADNI is greater among individuals with cognitive impairment and has been associated with AD neurodegenerative markers and cerebral amyloidosis. The negative additive effects of cerebrovascular disease appear present, even in select populations, and future biomarker work needs to further explore this relationship.
Collapse
Affiliation(s)
- Pauline Maillard
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Evan Fletcher
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| | - Owen Carmichael
- Biomedical Imaging, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | | | - Stephan Seiler
- Department of Neurology, University of California at Davis, Sacramento, California, USA
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Charles DeCarli
- Department of Neurology, University of California at Davis, Sacramento, California, USA
| |
Collapse
|
13
|
Sanchez-Martinez Y, Lopez-Lopez JP, Gomez-Montoya I, Hernandez-Quiñones D, Ruiz-Uribe G, Rincón-Rueda Z, Garcia RG, Lopez-Jaramillo P. Muscular strength, endothelial function and cognitive disorders: state of the art. J Physiol 2024. [PMID: 39612371 DOI: 10.1113/jp285939] [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: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 12/01/2024] Open
Abstract
In recent years, the ageing population has increasingly grown. This process carries a range of pathophysiological changes involving alterations in the skeletal muscle, vascular endothelium and brain function, becoming an important risk factor for developing cognitive disorders and cardiovascular diseases. With ageing, there is a decrease in muscle mass and muscle strength, and a relationship between muscle strength decrease and cognitive decline has been shown. Lower handgrip strength has been linked to memory impairment, lower global cognitive function, decreased attention and reduced visuospatial abilities in the elderly, but understanding of the underlying mechanisms that explain the link between altered skeletal muscle function and structure, endothelial dysfunction, and the role of endothelial dysfunction in the onset of cognitive disorders has been scarcely explored. This review aims to detail the cellular and molecular mechanisms by which the progressive changes associated with ageing can alter healthy skeletal muscle and endothelial function, creating an environment of oxidative stress, inflammation and mitochondrial dysfunction. These changes can lead to reduced muscle strength, and the secretion of detrimental endothelial factors, resulting in endothelial dysfunction, blood-brain barrier disruption, and damage to neurons and microglia, ultimately accelerating the onset of cognitive disorders in the elderly. In addition, we aimed to describe the mechanisms that potentially explain how preserving muscular function with resistance training could prevent brain function deterioration, including the production of different factors that allow an improved endothelial function, haemodynamic parameters and brain plasticity, ultimately delaying the onset of cognitive impairment and chronic diseases.
Collapse
Affiliation(s)
| | - Jose P Lopez-Lopez
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | | | | | - Gabriela Ruiz-Uribe
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | - Zully Rincón-Rueda
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
| | - Ronald G Garcia
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Patricio Lopez-Jaramillo
- Masira Research Institute, Universidad de Santander (UDES), Bucaramanga, Colombia
- Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
14
|
Makkinejad N, Zanon Zotin MC, van den Brink H, Auger CA, vom Eigen KA, Iglesias JE, Greenberg SM, Perosa V, van Veluw SJ. Neuropathological Correlates of White Matter Hyperintensities in Cerebral Amyloid Angiopathy. J Am Heart Assoc 2024; 13:e035744. [PMID: 39526350 PMCID: PMC11681407 DOI: 10.1161/jaha.124.035744] [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: 04/23/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are frequently observed on magnetic resonance imaging (MRI) in patients with cerebral amyloid angiopathy (CAA). The neuropathological substrates that underlie WMHs in CAA are unclear, and it remains largely unexplored whether the different WMH distribution patterns associated with CAA (posterior confluent and subcortical multispot) reflect alternative pathophysiological mechanisms. METHODS AND RESULTS We performed a combined in vivo MRI-ex vivo MRI-neuropathological study in patients with definite CAA. Formalin-fixed hemispheres from 19 patients with CAA, most of whom also had in vivo MRI available, underwent 3T MRI, followed by standard neuropathological examination of the hemispheres and targeted neuropathological assessment of WMH patterns. Ex vivo WMH volume was independently associated with CAA severity (P=0.046) but not with arteriolosclerosis (P=0.743). In targeted neuropathological examination, compared with normal-appearing white matter, posterior confluent WMHs were associated with activated microglia (P=0.043) and clasmatodendrosis (P=0.031), a form of astrocytic injury. Trends were found for an association with white matter rarefaction (P=0.074) and arteriolosclerosis (P=0.094). An exploratory descriptive analysis suggested that the histopathological correlates of WMH multispots were similar to those underlying posterior confluent WMHs. CONCLUSIONS This study confirmed that vascular amyloid β severity in the cortex is significantly associated with WMH volume in patients with definite CAA. The histopathological substrates of both posterior confluent and WMH multispots were comparable, suggesting overlapping pathophysiological mechanisms, although these exploratory observations require confirmation in larger studies.
Collapse
Affiliation(s)
- Nazanin Makkinejad
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Maria Clara Zanon Zotin
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
- Center for Imaging Sciences and Medical Physics, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoSPBrazil
| | - Hilde van den Brink
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Corinne A. Auger
- Department of NeurologyMassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical SchoolCharlestownMAUSA
| | - Kali A. vom Eigen
- Department of NeurologyMassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical SchoolCharlestownMAUSA
| | - Juan Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMAUSA
- Computer Science and Artificial Intelligence Laboratory, MITCambridgeMAUSA
- Centre for Medical Image ComputingUniversity College LondonLondonUnited Kingdom
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Valentina Perosa
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
| | - Susanne J. van Veluw
- J. Philip Kistler Stroke Research Center, Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMAUSA
- Department of NeurologyMassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical SchoolCharlestownMAUSA
| |
Collapse
|
15
|
Anfray A, Schaeffer S, Hattori Y, Santisteban MM, Casey N, Wang G, Strickland M, Zhou P, Holtzman DM, Anrather J, Park L, Iadecola C. A cell-autonomous role for border-associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. Nat Neurosci 2024; 27:2138-2151. [PMID: 39294490 PMCID: PMC11758676 DOI: 10.1038/s41593-024-01757-6] [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: 07/31/2023] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Apolipoprotein E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4, we report that border-associated macrophages (BAMs), myeloid cells closely apposed to neocortical microvessels, are both sources and effectors of ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAMs is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell-autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
- Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Samantha Schaeffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Monica M Santisteban
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Rahmani M, Dierker D, Yaeger L, Saykin A, Luckett PH, Vlassenko AG, Owens C, Jafri H, Womack K, Fripp J, Xia Y, Tosun D, Benzinger TLS, Masters CL, Lee JM, Morris JC, Goyal MS, Strain JF, Kukull W, Weiner M, Burnham S, CoxDoecke TJ, Fedyashov V, Fripp J, Shishegar R, Xiong C, Marcus D, Raniga P, Li S, Aschenbrenner A, Hassenstab J, Lim YY, Maruff P, Sohrabi H, Robertson J, Markovic S, Bourgeat P, Doré V, Mayo CJ, Mussoumzadeh P, Rowe C, Villemagne V, Bateman R, Fowler C, Li QX, Martins R, Schindler S, Shaw L, Cruchaga C, Harari O, Laws S, Porter T, O'Brien E, Perrin R, Kukull W, Bateman R, McDade E, Jack C, Morris J, Yassi N, Bourgeat P, Perrin R, Roberts B, Villemagne V, Fedyashov V, Goudey B. Evolution of white matter hyperintensity segmentation methods and implementation over the past two decades; an incomplete shift towards deep learning. Brain Imaging Behav 2024; 18:1310-1322. [PMID: 39083144 PMCID: PMC11582091 DOI: 10.1007/s11682-024-00902-w] [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] [Accepted: 06/26/2024] [Indexed: 08/22/2024]
Abstract
This systematic review examines the prevalence, underlying mechanisms, cohort characteristics, evaluation criteria, and cohort types in white matter hyperintensity (WMH) pipeline and implementation literature spanning the last two decades. Following Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, we categorized WMH segmentation tools based on their methodologies from January 1, 2000, to November 18, 2022. Inclusion criteria involved articles using openly available techniques with detailed descriptions, focusing on WMH as a primary outcome. Our analysis identified 1007 visual rating scales, 118 pipeline development articles, and 509 implementation articles. These studies predominantly explored aging, dementia, psychiatric disorders, and small vessel disease, with aging and dementia being the most prevalent cohorts. Deep learning emerged as the most frequently developed segmentation technique, indicative of a heightened scrutiny in new technique development over the past two decades. We illustrate observed patterns and discrepancies between published and implemented WMH techniques. Despite increasingly sophisticated quantitative segmentation options, visual rating scales persist, with the SPM technique being the most utilized among quantitative methods and potentially serving as a reference standard for newer techniques. Our findings highlight the need for future standards in WMH segmentation, and we provide recommendations based on these observations.
Collapse
Affiliation(s)
- Maryam Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Donna Dierker
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Andrew Saykin
- Department School of Medicine, Indiana University, Bloomington, IN, USA
| | - Patrick H Luckett
- Division of Neurotechnology, Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrei G Vlassenko
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, St. Louis, MO, USA
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher Owens
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hussain Jafri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle Womack
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jurgen Fripp
- The Australian E-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Ying Xia
- The Australian E-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Duygu Tosun
- Division of Radiology and Biomedical Imaging, University of CA - San Francisco, San Francisco, CA, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, St. Louis, MO, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer Disease Research Center, St. Louis, MO, USA
| | - Manu S Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy F Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Morrison C, Oliver MD, Kamal F, Dadar M. Beyond Hypertension: Examining Variable Blood Pressure's Role in Cognition and Brain Structure. J Gerontol B Psychol Sci Soc Sci 2024; 79:gbae121. [PMID: 39012223 PMCID: PMC11308164 DOI: 10.1093/geronb/gbae121] [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: 03/04/2024] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVES Hypertension or high blood pressure (BP) is one of the 12 modifiable risk factors that contribute to 40% of dementia cases that could be delayed or prevented. Although hypertension is associated with cognitive decline and structural brain changes, less is known about the long-term association between variable BP and cognitive/brain changes. This study examined the relationship between variable BP and longitudinal cognitive, white matter hyperintensity (WMH), gray matter (GM), and white matter (WM) volume change over time and postmortem neuropathology. METHODS A total of 4,606 participants (32,776 follow-ups) from RADC Research Resource Sharing Hub (RUSH) and 2,114 participants (9,827 follow-ups) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were included. Participants were divided into 1 of 3 groups: normal, high, or variable BP. Linear-mixed models investigated the relationship between BP and cognition, brain structure, and neuropathology. RESULTS Older adults with variable BP exhibited the highest rate of cognitive decline followed by high and then normal BP. Increased GM volume loss and WMH burden were also observed in variable compared to high and normal BP. In postmortem neuropathology, both variable and high BP had increased rates compared to normal BP. Results were consistent across the RUSH and ADNI participants, supporting the generalizability of the findings. DISCUSSION Damages potentially associated with variable BP may reduce resilience to future dementia-related pathology and increased the risk of dementia more than that caused by high BP. Improved treatment and management of variable BP may help reduce cognitive decline in the older adult population.
Collapse
Affiliation(s)
| | - Michael D Oliver
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee, USA
- Belmont Data Collaborative, Belmont University, Nashville, Tennessee, USA
| | - Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| |
Collapse
|
18
|
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer's disease. Acta Neuropathol 2024; 148:31. [PMID: 39196440 DOI: 10.1007/s00401-024-02790-2] [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: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
19
|
Björnfot C, Eklund A, Larsson J, Hansson W, Birnefeld J, Garpebring A, Qvarlander S, Koskinen LOD, Malm J, Wåhlin A. Cerebral arterial stiffness is linked to white matter hyperintensities and perivascular spaces in older adults - A 4D flow MRI study. J Cereb Blood Flow Metab 2024; 44:1343-1351. [PMID: 38315044 PMCID: PMC11342729 DOI: 10.1177/0271678x241230741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024]
Abstract
White matter hyperintensities (WMH), perivascular spaces (PVS) and lacunes are common MRI features of small vessel disease (SVD). However, no shared underlying pathological mechanism has been identified. We investigated whether SVD burden, in terms of WMH, PVS and lacune status, was related to changes in the cerebral arterial wall by applying global cerebral pulse wave velocity (gcPWV) measurements, a newly described marker of cerebral vascular stiffness. In a population-based cohort of 190 individuals, 66-85 years old, SVD features were estimated from T1-weighted and FLAIR images while gcPWV was estimated from 4D flow MRI data. Additionally, the gcPWV's stability to variations in field-of-view was analyzed. The gcPWV was 10.82 (3.94) m/s and displayed a significant correlation to WMH and white matter PVS volume (r = 0.29, p < 0.001; r = 0.21, p = 0.004 respectively from nonparametric tests) that persisted after adjusting for age, blood pressure variables, body mass index, ApoB/A1 ratio, smoking as well as cerebral pulsatility index, a previously suggested early marker of SVD. The gcPWV displayed satisfactory stability to field-of-view variations. Our results suggest that SVD is accompanied by changes in the cerebral arterial wall that can be captured by considering the velocity of the pulse wave transmission through the cerebral arterial network.
Collapse
Affiliation(s)
- Cecilia Björnfot
- Department of Diagnostics and Intervention, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Anders Eklund
- Department of Diagnostics and Intervention, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
| | - Jenny Larsson
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - William Hansson
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Johan Birnefeld
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Garpebring
- Department of Diagnostics and Intervention, Umeå University, Umeå, Sweden
| | - Sara Qvarlander
- Department of Diagnostics and Intervention, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Lars-Owe D Koskinen
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Jan Malm
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Wåhlin
- Department of Diagnostics and Intervention, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå, Sweden
- Department of Applied Physics and Electronics, Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Malla S, Bryant AG, Jayakumar R, Woost B, Wolf N, Li A, Das S, van Veluw SJ, Bennett RE. Molecular profiling of frontal and occipital subcortical white matter hyperintensities in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598845. [PMID: 38915516 PMCID: PMC11195168 DOI: 10.1101/2024.06.13.598845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
White matter hyperintensities (WMHs) are commonly detected on T2-weighted magnetic resonance imaging (MRI) scans, occurring in both typical aging and Alzheimer's disease. Despite their frequent appearance and their association with cognitive decline, the molecular factors contributing to WMHs remain unclear. In this study, we investigated the transcriptomic profiles of two commonly affected brain regions with coincident AD pathology-frontal subcortical white matter (frontal-WM) and occipital subcortical white matter (occipital-WM)-and compared with age-matched healthy controls. Through RNA-sequencing in frontal- and occipital-WM bulk tissues, we identified an upregulation of genes associated with brain vasculature function in AD white matter. To further elucidate vasculature-specific transcriptomic features, we performed RNA-seq analysis on blood vessels isolated from these white matter regions, which revealed an upregulation of genes related to protein folding pathways. Finally, comparing gene expression profiles between AD individuals with high- versus low-WMH burden showed an increased expression of pathways associated with immune function. Taken together, our study characterizes the diverse molecular profiles of white matter changes in AD compared to normal aging and provides new mechanistic insights processes underlying AD-related WMHs.
Collapse
Affiliation(s)
- Sulochan Malla
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Annie G Bryant
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- School of Physics, The University of Sydney, Sydney, Australia
| | - Rojashree Jayakumar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Benjamin Woost
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Nina Wolf
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Andrew Li
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sudeshna Das
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rachel E Bennett
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Chu M, Jiang D, Nan H, Wen L, Liu L, Qu M, Wu L. Vascular dysfunction in sporadic bvFTD: white matter hyperintensity and peripheral vascular biomarkers. Alzheimers Res Ther 2024; 16:72. [PMID: 38581060 PMCID: PMC10998369 DOI: 10.1186/s13195-024-01422-x] [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: 07/13/2023] [Accepted: 02/28/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND Vascular dysfunction was recently reported to be involved in the pathophysiological process of neurodegenerative diseases, but its role in sporadic behavioral variant frontotemporal dementia (bvFTD) remains unclear. The aim of this study was to systematically explore vascular dysfunction, including changes in white matter hyperintensities (WMHs) and peripheral vascular markers in bvFTD. METHODS Thirty-two patients with bvFTD who with no vascular risk factors were enrolled in this cross-sectional study and assessed using positron emission tomography/magnetic resonance (PET/MRI) imaging, peripheral plasma vascular/inflammation markers, and neuropsychological examinations. Group differences were tested using Student's t-tests and Mann-Whitney U tests. A partial correlation analysis was implemented to explore the association between peripheral vascular markers, neuroimaging, and clinical measures. RESULTS WMH was mainly distributed in anterior brain regions. All peripheral vascular factors including matrix metalloproteinases-1 (MMP-1), MMP-3, osteopontin, and pentraxin-3 were increased in the bvFTD group. WMH was associated with the peripheral vascular factor pentraxin-3. The plasma level of MMP-1 was negatively correlated with the gray matter metabolism of the frontal, temporal, insula, and basal ganglia brain regions. The WMHs in the frontal and limbic lobes were associated with plasma inflammation markers, disease severity, executive function, and behavior abnormality. Peripheral vascular markers were associated with the plasma inflammation markers. CONCLUSIONS WMHs and abnormalities in peripheral vascular markers were found in patients with bvFTD. These were found to be associated with the disease-specific pattern of neurodegeneration, indicating that vascular dysfunction may be involved in the pathogenesis of bvFTD. This warrants further confirmation by postmortem autopsy. Targeting the vascular pathway might be a promising approach for potential therapy.
Collapse
Affiliation(s)
- Min Chu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Deming Jiang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Haitian Nan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Lulu Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Li Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Miao Qu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Liyong Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
22
|
Coomans EM, van Westen D, Binette AP, Strandberg O, Spotorno N, Serrano GE, Beach TG, Palmqvist S, Stomrud E, Ossenkoppele R, Hansson O. Interactions between vascular burden and amyloid-β pathology on trajectories of tau accumulation. Brain 2024; 147:949-960. [PMID: 37721482 PMCID: PMC10907085 DOI: 10.1093/brain/awad317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/02/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023] Open
Abstract
Cerebrovascular pathology often co-exists with Alzheimer's disease pathology and can contribute to Alzheimer's disease-related clinical progression. However, the degree to which vascular burden contributes to Alzheimer's disease pathological progression is still unclear. This study aimed to investigate interactions between vascular burden and amyloid-β pathology on both baseline tau tangle load and longitudinal tau accumulation. We included 1229 participants from the Swedish BioFINDER-2 Study, including cognitively unimpaired and impaired participants with and without biomarker-confirmed amyloid-β pathology. All underwent baseline tau-PET (18F-RO948), and a subset (n = 677) underwent longitudinal tau-PET after 2.5 ± 1.0 years. Tau-PET uptake was computed for a temporal meta-region-of-interest. We focused on four main vascular imaging features and risk factors: microbleeds; white matter lesion volume; stroke-related events (infarcts, lacunes and haemorrhages); and the Framingham Heart Study Cardiovascular Disease risk score. To validate our in vivo results, we examined 1610 autopsy cases from an Arizona-based neuropathology cohort on three main vascular pathological features: cerebral amyloid angiopathy; white matter rarefaction; and infarcts. For the in vivo cohort, primary analyses included age-, sex- and APOE ɛ4-corrected linear mixed models between tau-PET (outcome) and interactions between time, amyloid-β and each vascular feature (predictors). For the neuropathology cohort, age-, sex- and APOE ɛ4-corrected linear models between tau tangle density (outcome) and an interaction between plaque density and each vascular feature (predictors) were performed. In cognitively unimpaired individuals, we observed a significant interaction between microbleeds and amyloid-β pathology on greater baseline tau load (β = 0.68, P < 0.001) and longitudinal tau accumulation (β = 0.11, P < 0.001). For white matter lesion volume, we did not observe a significant independent interaction effect with amyloid-β on tau after accounting for microbleeds. In cognitively unimpaired individuals, we further found that stroke-related events showed a significant negative interaction with amyloid-β on longitudinal tau (β = -0.08, P < 0.001). In cognitively impaired individuals, there were no significant interaction effects between cerebrovascular and amyloid-β pathology at all. In the neuropathology dataset, the in vivo observed interaction effects between cerebral amyloid angiopathy and plaque density (β = 0.38, P < 0.001) and between infarcts and plaque density (β = -0.11, P = 0.005) on tau tangle density were replicated. To conclude, we demonstrated that cerebrovascular pathology-in the presence of amyloid-β pathology-modifies tau accumulation in early stages of Alzheimer's disease. More specifically, the co-occurrence of microbleeds and amyloid-β pathology was associated with greater accumulation of tau aggregates during early disease stages. This opens the possibility that interventions targeting microbleeds may attenuate the rate of tau accumulation in Alzheimer's disease.
Collapse
Affiliation(s)
- Emma M Coomans
- Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081HV Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain Imaging, 1081HV Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081HV Amsterdam, The Netherlands
| | - Danielle van Westen
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081HV Amsterdam, The Netherlands
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
- Amsterdam Neuroscience, Neurodegeneration, 1071HV Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, SE-222 42 Lund, Sweden
- Memory Clinic, Skåne University Hospital, SE-205 02 Malmö, Sweden
| |
Collapse
|
23
|
Chen Y, Lu P, Wu S, Yang J, Liu W, Zhang Z, Xu Q. CD163-Mediated Small-Vessel Injury in Alzheimer's Disease: An Exploration from Neuroimaging to Transcriptomics. Int J Mol Sci 2024; 25:2293. [PMID: 38396970 PMCID: PMC10888773 DOI: 10.3390/ijms25042293] [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: 02/10/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Patients with Alzheimer's disease (AD) often present with imaging features indicative of small-vessel injury, among which, white-matter hyperintensities (WMHs) are the most prevalent. However, the underlying mechanism of the association between AD and small-vessel injury is still obscure. The aim of this study is to investigate the mechanism of small-vessel injury in AD. Differential gene expression analyses were conducted to identify the genes related to WMHs separately in mild cognitive impairment (MCI) and cognitively normal (CN) subjects from the ADNI database. The WMH-related genes identified in patients with MCI were considered to be associated with small-vessel injury in early AD. Functional enrichment analyses and a protein-protein interaction (PPI) network were performed to explore the pathway and hub genes related to the mechanism of small-vessel injury in MCI. Subsequently, the Boruta algorithm and support vector machine recursive feature elimination (SVM-RFE) algorithm were performed to identify feature-selection genes. Finally, the mechanism of small-vessel injury was analyzed in MCI from the immunological perspectives; the relationship of feature-selection genes with various immune cells and neuroimaging indices were also explored. Furthermore, 5×FAD mice were used to demonstrate the genes related to small-vessel injury. The results of the logistic regression analyses suggested that WMHs significantly contributed to MCI, the early stage of AD. A total of 276 genes were determined as WMH-related genes in patients with MCI, while 203 WMH-related genes were obtained in CN patients. Among them, only 15 genes overlapped and were thus identified as the crosstalk genes. By employing the Boruta and SVM-RFE algorithms, CD163, ALDH3B1, MIR22HG, DTX2, FOLR2, ALDH2, and ZNF23 were recognized as the feature-selection genes linked to small-vessel injury in MCI. After considering the results from the PPI network, CD163 was finally determined as the critical WMH-related gene in MCI. The expression of CD163 was correlated with fractional anisotropy (FA) values in regions that are vulnerable to small-vessel injury in AD. The immunostaining and RT-qPCR results from the verifying experiments demonstrated that the indicators of small-vessel injury presented in the cortical tissue of 5×FAD mice and related to the upregulation of CD163 expression. CD163 may be the most pivotal candidates related to small-vessel injury in early AD.
Collapse
Affiliation(s)
- Yuewei Chen
- Health Management Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.C.); (P.L.); (W.L.)
- Department of Neurology, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Peiwen Lu
- Health Management Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.C.); (P.L.); (W.L.)
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shengju Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jie Yang
- Health Management Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.C.); (P.L.); (W.L.)
- Department of Neurology, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wanwan Liu
- Health Management Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.C.); (P.L.); (W.L.)
| | - Zhijun Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qun Xu
- Health Management Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.C.); (P.L.); (W.L.)
- Department of Neurology, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital of Medical School, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
24
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [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: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
25
|
Ly MT, Tuz-Zahra F, Tripodis Y, Adler CH, Balcer LJ, Bernick C, Zetterberg H, Blennow K, Peskind ER, Au R, Banks SJ, Barr WB, Wethe JV, Bondi MW, Delano-Wood LM, Cantu RC, Coleman MJ, Dodick DW, McClean MD, Mez JB, Palmisano J, Martin B, Hartlage K, Lin AP, Koerte IK, Cummings JL, Reiman EM, Shenton ME, Stern RA, Bouix S, Alosco ML. Association of Vascular Risk Factors and CSF and Imaging Biomarkers With White Matter Hyperintensities in Former American Football Players. Neurology 2024; 102:e208030. [PMID: 38165330 PMCID: PMC10870736 DOI: 10.1212/wnl.0000000000208030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/13/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Recent data link exposure to repetitive head impacts (RHIs) from American football with increased white matter hyperintensity (WMH) burden. WMH might have unique characteristics in the context of RHI beyond vascular risk and normal aging processes. We evaluated biological correlates of WMH in former American football players, including markers of amyloid, tau, inflammation, axonal injury, neurodegeneration, and vascular health. METHODS Participants underwent clinical interviews, MRI, and lumbar puncture as part of the Diagnostics, Imaging, and Genetics Network for the Objective Study and Evaluation of Chronic Traumatic Encephalopathy Research Project. Structural equation modeling tested direct and indirect effects between log-transformed total fluid-attenuated inversion recovery (FLAIR) lesion volumes (TLV) and the revised Framingham stroke risk profile (rFSRP), MRI-derived global metrics of cortical thickness and fractional anisotropy (FA), and CSF levels of amyloid β1-42, p-tau181, soluble triggering receptor expressed on myeloid cells 2 (sTREM2), and neurofilament light. Covariates included age, race, education, body mass index, APOE ε4 carrier status, and evaluation site. Models were performed separately for former football players and a control group of asymptomatic men unexposed to RHI. RESULTS In 180 former football players (mean age = 57.2, 36% Black), higher log(TLV) had direct associations with the following: higher rFSRP score (B = 0.26, 95% CI 0.07-0.40), higher p-tau181 (B = 0.17, 95% CI 0.01-0.43), lower FA (B = -0.28, 95% CI -0.42 to -0.13), and reduced cortical thickness (B = -0.25, 95% CI -0.45 to -0.08). In 60 asymptomatic unexposed men (mean age = 59.3, 40% Black), there were no direct effects on log(TLV) (rFSRP: B = -0.03, 95% CI -0.48 to 0.57; p-tau181: B = -0.30, 95% CI -1.14 to 0.37; FA: B = -0.07, 95% CI -0.48 to 0.42; or cortical thickness: B = -0.28, 95% CI -0.64 to 0.10). The former football players showed stronger associations between log(TLV) and rFSRP (1,069% difference in estimates), p-tau181 (158%), and FA (287%) than the unexposed men. DISCUSSION Risk factors and biological correlates of WMH differed between former American football players and asymptomatic unexposed men. In addition to vascular health, p-tau181 and diffusion tensor imaging indices of white matter integrity showed stronger associations with WMH in the former football players. FLAIR WMH may have specific risk factors and pathologic underpinnings in RHI-exposed individuals.
Collapse
Affiliation(s)
- Monica T Ly
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Fatima Tuz-Zahra
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Yorghos Tripodis
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Charles H Adler
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Laura J Balcer
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Charles Bernick
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Henrik Zetterberg
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Kaj Blennow
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Elaine R Peskind
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Rhoda Au
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Sarah J Banks
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - William B Barr
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Jennifer V Wethe
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Mark W Bondi
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Lisa M Delano-Wood
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Robert C Cantu
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Michael J Coleman
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - David W Dodick
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Michael D McClean
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Jesse B Mez
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Joseph Palmisano
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Brett Martin
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Kaitlin Hartlage
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Alexander P Lin
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Inga K Koerte
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Jeffrey L Cummings
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Eric M Reiman
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Martha E Shenton
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Robert A Stern
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Sylvain Bouix
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| | - Michael L Alosco
- From the VA San Diego Healthcare System (M.T.L., M.W.B., L.M.D.-W.), CA; Department of Psychiatry (M.T.L., S.J.B., M.W.B., L.M.D.-W.), University of California San Diego Health, La Jolla; Departments of Biostatistics (F.T.-Z., Y.T.), Epidemiology (R.A.), Environmental Health (M.D.M.), Biostatistics and Epidemiology Data Analytics Center (J.P., B.M., K.H.), Boston University School of Public Health, MA; Boston University Alzheimer's Disease Research Center (Y.T., J.B.M., M.L.A., R.A., R.C.C., R.A.S.), Boston University CTE Center; Department of Neurology, Boston University Chobanian & Avedisian School of Medicine; Departments of Neurology (C.H.A., D.W.D.) and Psychiatry and Psychology (J.V.W.), Mayo Clinic School of Medicine, Mayo Clinic Arizona, Scottsdale; Departments of Neurology (L.J.B.), Population Health and Ophthalmology, (L.J.B.), and Neurology (W.B.B.), NYU Grossman School of Medicine; Cleveland Clinic Lou Ruvo Center for Brain Health (C.B.), Las Vegas, NV; Department of Neurology (C.B.), University of Washington, Seattle; Department of Neurodegenerative Disease (H.Z.), and UK Dementia Research Institute (H.Z.), University College London Institute of Neurology, UK; Hong Kong Center for Neurodegenerative Diseases (H.Z.), China; Wisconsin Alzheimer's Disease Research Center (H.Z.), University of Wisconsin-Madison; Clinical Neurochemistry Laboratory (H.Z., K.B.), Sahlgrenska University Hospital, Gothenburg; Department of Psychiatry and Neurochemistry (K.B.), Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Sweden; VA Northwest Mental Illness Research, Education, and Clinical Center (E.R.P.), Seattle, WA; Department of Psychiatry and Behavioral Sciences (E.R.P.), University of Washington School of Medicine, Seattle; Framingham Heart Study (R.A., J.B.M.); Slone Epidemiology Center (R.A.), Boston University, MA; Department of Neurosciences (S.J.B.), University of California San Diego; Psychiatry Neuroimaging Laboratory (M.J.C., A.P.L., I.K.K., M.E.S., S.B.), Departments of Psychiatry Radiology (M.E.S.), and Center for Clinical Spectroscopy (A.P.L.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; cBRAIN (I.K.K.), Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, Ludwigs-Maximilians-Universität, Munich, Germany; Chambers-Grundy Center for Transformative Neuroscience (J.L.C.), Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas; Banner Alzheimer's Institute (E.M.R.), Phoenix; Department of Psychiatry (E.M.R.), University of Arizona, Phoenix; Arizona State University (E.M.R.), Phoenix; Translational Genomics Research Institute (E.M.R.), Phoenix; Arizona Alzheimer's Consortium (E.M.R.), Phoenix; and Department of Software Engineering and Information Technology (S.B.), École de technologie supérieure, Université du Québec, Montréal, Canada
| |
Collapse
|
26
|
Morrison C, Oliver MD, Kamal F, Dadar M. Beyond Hypertension: Examining Variable Blood Pressure's Role in Cognition and Brain Structure. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.15.24301335. [PMID: 38293179 PMCID: PMC10827268 DOI: 10.1101/2024.01.15.24301335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Importance Hypertension is a known risk factor for cognitive decline and structural brain changes in aging and dementia. In addition to high blood pressure (BP), individuals may also experience variable BP, meaning that their BP fluctuates between normal and high. It is currently unclear what the effects of variable BP are on cognition and brain structure. Objective To investigate the influence of BP on cognition and brain structure in older adults. Design Setting and Participants This longitudinal cohort study included data from the Rush Alzheimer's Disease Center Research Resource Sharing Hub (RUSH) and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants from the two studies were included if they had BP measurements and either cognitive scores or MRI scans from at least one visit. Main Outcomes and Measures Longitudinal gray matter, white matter, white matter hyperintensity volumes, postmortem neuropathology information, as well as cognitive test scores. Results A total of 4606 participants (3429 females, mean age = 76.8) with 32776 follow-ups (mean = 7 years) from RUSH and 2114 participants (1132 females, mean age = 73.3) with 9827 follow-ups (mean = 3 years) from ADNI were included in this study. Participants were divided into one of three groups: 1) normal BP, high BP, or variable BP. Older adults with variable BP exhibited the highest rate of cognitive decline followed by high BP and then normal BP. Increased GM volume loss and WMH burden was also observed in variable BP compared to high and normal BP. With respect to post-mortem neuropathology, both variable and high BP had increased severities compared to normal BP. Importantly, results were consistent across the RUSH and ADNI participants, supporting the generalizability of the findings. Conclusion and Relevance Limited research has examined the long-term impact of variable BP on cognition and brain structure. These findings show the importance that both high and variable BP have on cognitive decline and structural brain changes. Structural damages caused by variable BP may reduce resilience to future dementia-related pathology and increased risk of dementia. Improved treatment and management of variable BP may help reduce cognitive decline in the older adult population.
Collapse
Affiliation(s)
| | - Michael D Oliver
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee, United States
- Belmont Data Collaborative, Belmont University, Nashville, Tennessee, United States
| | - Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| |
Collapse
|
27
|
Mueller SG. Traumatic Brain Injury and Post-Traumatic Stress Disorder and Their Influence on Development and Pattern of Alzheimer's Disease Pathology in Later Life. J Alzheimers Dis 2024; 98:1427-1441. [PMID: 38552112 DOI: 10.3233/jad-231183] [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] [Indexed: 04/20/2024]
Abstract
Background Traumatic brain injury (TBI) and posttraumatic stress disorder (PTSD) are potential risk factors for the development of dementia including Alzheimer's disease (AD) in later life. The findings of studies investigating this question are inconsistent though. Objective To investigate if these inconsistencies are caused by the existence of subgroups with different vulnerability for AD pathology and if these subgroups are characterized by atypical tau load/atrophy pattern. Methods The MRI and PET data of 89 subjects with or without previous TBI and/or PTSD from the DoD ADNI database were used to calculate an age-corrected gray matter tau mismatch metric (ageN-T mismatch-score and matrix) for each subject. This metric provides a measure to what degree regional tau accumulation drives regional gray matter atrophy (matrix) and can be used to calculate a summary score (score) reflecting the severity of AD pathology in an individual. Results The ageN-T mismatch summary score was positively correlated with whole brain beta-amyloid load and general cognitive function but not with PTSD or TBI severity. Hierarchical cluster analysis identified five different spatial patterns of tau-gray matter interactions. These clusters reflected the different stages of the typical AD tau progression pattern. None was exclusively associated with PTSD and/or TBI. Conclusions These findings suggest that a) although subsets of patients with PTSD and/or TBI develop AD-pathology, a history of TBI or PTSD alone or both is not associated with a significantly higher risk to develop AD pathology in later life. b) remote TBI or PTSD do not modify the typical AD pathology distribution pattern.
Collapse
Affiliation(s)
- Susanne G Mueller
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
28
|
Strain JF, Rahmani M, Dierker D, Owen C, Jafri H, Vlassenko AG, Womack K, Fripp J, Tosun D, Benzinger TLS, Weiner M, Masters C, Lee JM, Morris JC, Goyal MS. Accuracy of TrUE-Net in comparison to established white matter hyperintensity segmentation methods: An independent validation study. Neuroimage 2024; 285:120494. [PMID: 38086495 PMCID: PMC11534282 DOI: 10.1016/j.neuroimage.2023.120494] [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: 06/28/2023] [Revised: 10/23/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
White matter hyperintensities (WMH) are nearly ubiquitous in the aging brain, and their topography and overall burden are associated with cognitive decline. Given their numerosity, accurate methods to automatically segment WMH are needed. Recent developments, including the availability of challenge data sets and improved deep learning algorithms, have led to a new promising deep-learning based automated segmentation model called TrUE-Net, which has yet to undergo rigorous independent validation. Here, we compare TrUE-Net to six established automated WMH segmentation tools, including a semi-manual method. We evaluated the techniques at both global and regional level to compare their ability to detect the established relationship between WMH burden and age. We found that TrUE-Net was highly reliable at identifying WMH regions with low false positive rates, when compared to semi-manual segmentation as the reference standard. TrUE-Net performed similarly or favorably when compared to the other automated techniques. Moreover, TrUE-Net was able to detect relationships between WMH and age to a similar degree as the reference standard semi-manual segmentation at both the global and regional level. These results support the use of TrUE-Net for identifying WMH at the global or regional level, including in large, combined datasets.
Collapse
Affiliation(s)
- Jeremy F Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis MO, USA.
| | - Maryam Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis MO, USA
| | - Donna Dierker
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis MO, USA
| | - Christopher Owen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hussain Jafri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrei G Vlassenko
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis MO, USA
| | - Kyle Womack
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, QLD, Australia
| | - Duygu Tosun
- Division of Radiology and Biomedical Imaging, University of California - San Francisco, San Francisco, CA, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer Disease Research Center, St. Louis, MO, USA
| | - Michael Weiner
- Division of Radiology and Biomedical Imaging, University of California - San Francisco, San Francisco, CA, USA
| | - Colin Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Knight Alzheimer Disease Research Center, St. Louis, MO, USA
| | - Manu S Goyal
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Neuroimaging Labs Research Center, Washington University School of Medicine, St. Louis MO, USA
| |
Collapse
|
29
|
Zilioli A, Misirocchi F, Pancaldi B, Mutti C, Ganazzoli C, Morelli N, Pellegrini FF, Messa G, Scarlattei M, Mohanty R, Ruffini L, Westman E, Spallazzi M. Predicting amyloid-PET status in a memory clinic: The role of the novel antero-posterior index and visual rating scales. J Neurol Sci 2023; 455:122806. [PMID: 38006829 DOI: 10.1016/j.jns.2023.122806] [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: 09/18/2023] [Revised: 10/27/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
INTRODUCTION Visual rating scales are increasingly utilized in clinical practice to assess atrophy in crucial brain regions among patients with cognitive disorders. However, their capacity to predict Alzheimer's disease (AD)-related pathology remains unexplored, particularly within a heterogeneous memory clinic population. This study aims to assess the accuracy of a novel visual rating assessment, the antero-posterior index (API) scale, in predicting amyloid-PET status. Furthermore, the study seeks to determine the optimal cohort-based cutoffs for the medial temporal atrophy (MTA) and parietal atrophy (PA) scales and to integrate the main visual rating scores into a predictive model. METHODS We conducted a retrospective analysis of brain MRI and high-resolution TC scans from 153 patients with cognitive disorders who had undergone amyloid-PET assessments due to suspected AD pathology in a real-world memory clinic setting. RESULTS The API scale (cutoff ≥1) exhibited the highest accuracy (AUC = 0.721) among the visual rating scales. The combination of the cohort-based MTA and PA threshold with the API yielded favorable accuracy (AUC = 0.787). Analyzing a cohort of MCI/Mild dementia patients below 75 years of age, the API scale and the predictive model improved their accuracy (AUC = 0.741 and 0.813, respectively), achieving excellent results in the early-onset population (AUC = 0.857 and 0.949, respectively). CONCLUSION Our study emphasizes the significance of visual rating scales in predicting amyloid-PET positivity within a real-world memory clinic. Implementing the novel API scale, alongside our cohort-based MTA and PA thresholds, has the potential to substantially enhance diagnostic accuracy.
Collapse
Affiliation(s)
- Alessandro Zilioli
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Francesco Misirocchi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy.
| | - Beatrice Pancaldi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Carlotta Mutti
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy; Sleep Disorders Center, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Nicola Morelli
- Department of Neurology, G. da Saliceto Hospital, Piacenza, Italy
| | | | - Giovanni Messa
- Center for Cognitive Disorders, AUSL Parma, Parma, Italy
| | - Maura Scarlattei
- Nuclear Medicine Unit, University Hospital of Parma, Parma, Italy
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
| | - Livia Ruffini
- Nuclear Medicine Unit, University Hospital of Parma, Parma, Italy
| | - Eric Westman
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden; Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marco Spallazzi
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
| |
Collapse
|
30
|
Shirzadi Z, Schultz SA, Yau WYW, Joseph-Mathurin N, Fitzpatrick CD, Levin R, Kantarci K, Preboske GM, Jack CR, Farlow MR, Hassenstab J, Jucker M, Morris JC, Xiong C, Karch CM, Levey AI, Gordon BA, Schofield PR, Salloway SP, Perrin RJ, McDade E, Levin J, Cruchaga C, Allegri RF, Fox NC, Goate A, Day GS, Koeppe R, Chui HC, Berman S, Mori H, Sanchez-Valle R, Lee JH, Rosa-Neto P, Ruthirakuhan M, Wu CY, Swardfager W, Benzinger TLS, Sohrabi HR, Martins RN, Bateman RJ, Johnson KA, Sperling RA, Greenberg SM, Schultz AP, Chhatwal JP. Etiology of White Matter Hyperintensities in Autosomal Dominant and Sporadic Alzheimer Disease. JAMA Neurol 2023; 80:1353-1363. [PMID: 37843849 PMCID: PMC10580156 DOI: 10.1001/jamaneurol.2023.3618] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 10/17/2023]
Abstract
Importance Increased white matter hyperintensity (WMH) volume is a common magnetic resonance imaging (MRI) finding in both autosomal dominant Alzheimer disease (ADAD) and late-onset Alzheimer disease (LOAD), but it remains unclear whether increased WMH along the AD continuum is reflective of AD-intrinsic processes or secondary to elevated systemic vascular risk factors. Objective To estimate the associations of neurodegeneration and parenchymal and vessel amyloidosis with WMH accumulation and investigate whether systemic vascular risk is associated with WMH beyond these AD-intrinsic processes. Design, Setting, and Participants This cohort study used data from 3 longitudinal cohort studies conducted in tertiary and community-based medical centers-the Dominantly Inherited Alzheimer Network (DIAN; February 2010 to March 2020), the Alzheimer's Disease Neuroimaging Initiative (ADNI; July 2007 to September 2021), and the Harvard Aging Brain Study (HABS; September 2010 to December 2019). Main Outcome and Measures The main outcomes were the independent associations of neurodegeneration (decreases in gray matter volume), parenchymal amyloidosis (assessed by amyloid positron emission tomography), and vessel amyloidosis (evidenced by cerebral microbleeds [CMBs]) with cross-sectional and longitudinal WMH. Results Data from 3960 MRI sessions among 1141 participants were included: 252 pathogenic variant carriers from DIAN (mean [SD] age, 38.4 [11.2] years; 137 [54%] female), 571 older adults from ADNI (mean [SD] age, 72.8 [7.3] years; 274 [48%] female), and 318 older adults from HABS (mean [SD] age, 72.4 [7.6] years; 194 [61%] female). Longitudinal increases in WMH volume were greater in individuals with CMBs compared with those without (DIAN: t = 3.2 [P = .001]; ADNI: t = 2.7 [P = .008]), associated with longitudinal decreases in gray matter volume (DIAN: t = -3.1 [P = .002]; ADNI: t = -5.6 [P < .001]; HABS: t = -2.2 [P = .03]), greater in older individuals (DIAN: t = 6.8 [P < .001]; ADNI: t = 9.1 [P < .001]; HABS: t = 5.4 [P < .001]), and not associated with systemic vascular risk (DIAN: t = 0.7 [P = .40]; ADNI: t = 0.6 [P = .50]; HABS: t = 1.8 [P = .06]) in individuals with ADAD and LOAD after accounting for age, gray matter volume, CMB presence, and amyloid burden. In older adults without CMBs at baseline, greater WMH volume was associated with CMB development during longitudinal follow-up (Cox proportional hazards regression model hazard ratio, 2.63; 95% CI, 1.72-4.03; P < .001). Conclusions and Relevance The findings suggest that increased WMH volume in AD is associated with neurodegeneration and parenchymal and vessel amyloidosis but not with elevated systemic vascular risk. Additionally, increased WMH volume may represent an early sign of vessel amyloidosis preceding the emergence of CMBs.
Collapse
Affiliation(s)
- Zahra Shirzadi
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Stephanie A. Schultz
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Wai-Ying W. Yau
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | | | - Colleen D. Fitzpatrick
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Raina Levin
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Jason Hassenstab
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Tübingen, Germany
| | - John C. Morris
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Chengjie Xiong
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Celeste M. Karch
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | | | - Brian A. Gordon
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Peter R. Schofield
- Neuroscience Research Australia, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Richard J. Perrin
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Eric McDade
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, German Center for Neurodegenerative Diseases, site Munich, Munich Cluster for Systems Neurology, Munich, Germany
| | - Carlos Cruchaga
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | | | - Nick C. Fox
- UK Dementia Research Institute, University College London, London, United Kingdom
| | - Alison Goate
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor
| | - Helena C. Chui
- Keck School of Medicine, University of Southern California, Los Angeles
| | - Sarah Berman
- Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hiroshi Mori
- Osaka Metropolitan University Medical School, Osaka, Nagaoka Sutoku University, Osaka City, Niigata, Japan
| | | | - Jae-Hong Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Pedro Rosa-Neto
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Myuri Ruthirakuhan
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Che-Yuan Wu
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | | | - Hamid R. Sohrabi
- Centre for Healthy Ageing, School of Psychology, Health Future Institute, Murdoch University, Perth, Western Australia, Australia
| | - Ralph N. Martins
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Randall J. Bateman
- Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Keith A. Johnson
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Reisa A. Sperling
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Steven M. Greenberg
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Aaron P. Schultz
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| | - Jasmeer P. Chhatwal
- Massachusetts General Hospital, Brigham and Women’s Hospital, Harvard Medical School, Boston
| |
Collapse
|
31
|
Yu F, Iacono D, Perl DP, Lai C, Gill J, Le TQ, Lee P, Sukumar G, Armstrong RC. Neuronal tau pathology worsens late-phase white matter degeneration after traumatic brain injury in transgenic mice. Acta Neuropathol 2023; 146:585-610. [PMID: 37578550 PMCID: PMC10499978 DOI: 10.1007/s00401-023-02622-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Traumatic brain injury (TBI) causes diffuse axonal injury which can produce chronic white matter pathology and subsequent post-traumatic neurodegeneration with poor patient outcomes. Tau modulates axon cytoskeletal functions and undergoes phosphorylation and mis-localization in neurodegenerative disorders. The effects of tau pathology on neurodegeneration after TBI are unclear. We used mice with neuronal expression of human mutant tau to examine effects of pathological tau on white matter pathology after TBI. Adult male and female hTau.P301S (Tg2541) transgenic and wild-type (Wt) mice received either moderate single TBI (s-TBI) or repetitive mild TBI (r-mTBI; once daily × 5), or sham procedures. Acutely, s-TBI produced more extensive axon damage in the corpus callosum (CC) as compared to r-mTBI. After s-TBI, significant CC thinning was present at 6 weeks and 4 months post-injury in Wt and transgenic mice, with homozygous tau expression producing additional pathology of late demyelination. In contrast, r-mTBI did not produce significant CC thinning except at the chronic time point of 4 months in homozygous mice, which exhibited significant CC atrophy (- 29.7%) with increased microgliosis. Serum neurofilament light quantification detected traumatic axonal injury at 1 day post-TBI in Wt and homozygous mice. At 4 months, high tau and neurofilament in homozygous mice implicated tau in chronic axon pathology. These findings did not have sex differences detected. Conclusions: Neuronal tau pathology differentially exacerbated CC pathology based on injury severity and chronicity. Ongoing CC atrophy from s-TBI became accompanied by late demyelination. Pathological tau significantly worsened CC atrophy during the chronic phase after r-mTBI.
Collapse
Affiliation(s)
- Fengshan Yu
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Diego Iacono
- Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Daniel P Perl
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Chen Lai
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Tuan Q Le
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Patricia Lee
- Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Defense-Uniformed Services University Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Gauthaman Sukumar
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Regina C Armstrong
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD, 20814, USA.
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
32
|
Padrela BE, Lorenzini L, Collij LE, García DV, Coomans E, Ingala S, Tomassen J, Deckers Q, Shekari M, de Geus EJC, van de Giessen E, Kate MT, Visser PJ, Barkhof F, Petr J, den Braber A, Mutsaerts HJMM. Genetic, vascular and amyloid components of cerebral blood flow in a preclinical population. J Cereb Blood Flow Metab 2023; 43:1726-1736. [PMID: 37231665 PMCID: PMC10581242 DOI: 10.1177/0271678x231178993] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/05/2023] [Accepted: 04/09/2023] [Indexed: 05/27/2023]
Abstract
Aging-related cognitive decline can be accelerated by a combination of genetic factors, cardiovascular and cerebrovascular dysfunction, and amyloid-β burden. Whereas cerebral blood flow (CBF) has been studied as a potential early biomarker of cognitive decline, its normal variability in healthy elderly is less known. In this study, we investigated the contribution of genetic, vascular, and amyloid-β components of CBF in a cognitively unimpaired (CU) population of monozygotic older twins. We included 134 participants who underwent arterial spin labeling (ASL) MRI and [18F]flutemetamol amyloid-PET imaging at baseline and after a four-year follow-up. Generalized estimating equations were used to investigate the associations of amyloid burden and white matter hyperintensities with CBF. We showed that, in CU individuals, CBF: 1) has a genetic component, as within-pair similarities in CBF values were moderate and significant (ICC > 0.40); 2) is negatively associated with cerebrovascular damage; and 3) is positively associated with the interaction between cardiovascular risk scores and early amyloid-β burden, which may reflect a vascular compensatory response of CBF to early amyloid-β accumulation. These findings encourage future studies to account for multiple interactions with CBF in disease trajectory analyses.
Collapse
Affiliation(s)
- Beatriz E Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Luigi Lorenzini
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Lyduine E Collij
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - David Vállez García
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Emma Coomans
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Quinten Deckers
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Mahnaz Shekari
- BBRC: Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Pompeu Fabra University, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Eco JC de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Mara ten Kate
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Henk JMM Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Strain JF, Phuah CL, Adeyemo B, Cheng K, Womack KB, McCarthy J, Goyal M, Chen Y, Sotiras A, An H, Xiong C, Scharf A, Newsom-Stewart C, Morris JC, Benzinger TLS, Lee JM, Ances BM. White matter hyperintensity longitudinal morphometric analysis in association with Alzheimer disease. Alzheimers Dement 2023; 19:4488-4497. [PMID: 37563879 PMCID: PMC10592317 DOI: 10.1002/alz.13377] [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: 11/29/2022] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Vascular damage in Alzheimer's disease (AD) has shown conflicting findings particularly when analyzing longitudinal data. We introduce white matter hyperintensity (WMH) longitudinal morphometric analysis (WLMA) that quantifies WMH expansion as the distance from lesion voxels to a region of interest boundary. METHODS WMH segmentation maps were derived from 270 longitudinal fluid-attenuated inversion recovery (FLAIR) ADNI images. WLMA was performed on five data-driven WMH patterns with distinct spatial distributions. Amyloid accumulation was evaluated with WMH expansion across the five WMH patterns. RESULTS The preclinical group had significantly greater expansion in the posterior ventricular WM compared to controls. Amyloid significantly associated with frontal WMH expansion primarily within AD individuals. WLMA outperformed WMH volume changes for classifying AD from controls primarily in periventricular and posterior WMH. DISCUSSION These data support the concept that localized WMH expansion continues to proliferate with amyloid accumulation throughout the entirety of the disease in distinct spatial locations.
Collapse
Affiliation(s)
- Jeremy Fuller Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chia-Ling Phuah
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Babatunde Adeyemo
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kathleen Cheng
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kyle B Womack
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John McCarthy
- Department of Mathematics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Manu Goyal
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yasheng Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hongyu An
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chengjie Xiong
- Division of Biostatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Scharf
- Department of Biological Sciences, Missouri University for Science and Technology, Rolla, Missouri, USA
| | - Catherine Newsom-Stewart
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Carl Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| | - Tammie Lee Smith Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Kamal F, Morrison C, Maranzano J, Zeighami Y, Dadar M. White Matter Hyperintensity Trajectories in Patients With Progressive and Stable Mild Cognitive Impairment. Neurology 2023; 101:e815-e824. [PMID: 37407262 PMCID: PMC10449435 DOI: 10.1212/wnl.0000000000207514] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/25/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES White matter hyperintensities (WMH) are pathologic brain changes that are associated with increased age and cognitive decline. However, the association of WMH burden with amyloid positivity and conversion to dementia in people with mild cognitive impairment (MCI) is unclear. The aim of this study was to expand on this research by examining whether change in WMH burden over time differs in amyloid-negative (Aβ-) and amyloid-positive (Aβ+) people with MCI who either remain stable or convert to dementia. To examine this question, we compared regional WMH burden in 4 groups: Aβ+ progressor, Aβ- progressor, Aβ+ stable, and Aβ- stable. METHODS Participants with MCI from the Alzheimer Disease Neuroimaging Initiative were included if they had APOE ɛ4 status and if amyloid measures were available to determine amyloid status (i.e., Aβ+, or Aβ-). Participants with a baseline diagnosis of MCI and who had APOE ɛ4 information and amyloid measures were included. An average of 5.7 follow-up time points per participant were included, with a total of 5,054 follow-up time points with a maximum follow-up duration of 13 years. Differences in total and regional WMH burden were examined using linear mixed-effects models. RESULTS A total of 820 participants (55-90 years of age) were included in the study (Aβ+ progressor, n = 239; Aβ- progressor, n = 22; Aβ+ stable, n = 343; Aβ- stable, n = 216). People who were Aβ- stable exhibited reduced baseline WMH compared with Aβ+ progressors and people who were Aβ+ stable at all regions of interest (β belongs to 0.20-0.33, CI belongs to 0.03-0.49, p < 0.02), except deep WMH. When examining longitudinal results, compared with people who were Aβ- stable, all groups had steeper accumulation in WMH burden with Aβ+ progressors (β belongs to -0.03 to 0.06, CI belongs to -0.05 to 0.09, p < 0.01) having the largest increase (i.e., largest increase in WMH accumulation over time). DISCUSSION These results indicate that WMH accumulation contributes to conversion to dementia in older adults with MCI who are Aβ+ and Aβ-.
Collapse
Affiliation(s)
- Farooq Kamal
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada.
| | - Cassandra Morrison
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Josefina Maranzano
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Yashar Zeighami
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Mahsa Dadar
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| |
Collapse
|
35
|
Sepulveda-Falla D, Lanau CAV, White C, Serrano GE, Acosta-Uribe J, Mejía-Cupajita B, Villalba-Moreno ND, Lu P, Glatzel M, Kofler JK, Ghetti B, Frosch MP, Restrepo FL, Kosik KS, Beach TG. Comorbidities in Early-Onset Sporadic versus Presenilin-1 Mutation-Associated Alzheimer's Disease Dementia: Evidence for Dependency on Alzheimer's Disease Neuropathological Changes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.14.23294081. [PMID: 37646002 PMCID: PMC10462216 DOI: 10.1101/2023.08.14.23294081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Autopsy studies have demonstrated that comorbid neurodegenerative and cerebrovascular disease occur in the great majority of subjects with Alzheimer disease dementia (ADD), and are likely to additively alter the rate of decline or severity of cognitive impairment. The most important of these are Lewy body disease (LBD), TDP-43 proteinopathy and cerebrovascular disease, including white matter rarefaction (WMR) and cerebral infarcts. Comorbidities may interfere with ADD therapeutic trials evaluation of ADD clinical trials as they may not respond to AD-specific molecular therapeutics. It is possible, however, that at least some comorbidities may be, to some degree, secondary consequences of AD pathology, and if this were true then effective AD-specific therapeutics might also reduce the extent or severity of comorbid pathology. Comorbidities in ADD caused by autosomal dominant mutations such as those in the presenilin-1 (PSEN1) gene may provide an advantageous perspective on their pathogenesis, and deserve attention because these subjects are increasingly being entered into clinical trials. As ADD associated with PSEN1 mutations has a presumed single-cause etiology, and the average age at death is under 60, any comorbidities in this setting may be considered as at least partially secondary to the causative AD mechanisms rather than aging, and thus indicate whether effective ADD therapeutics may also be effective for comorbidities. In this study, we sought to compare the rates and types of ADD comorbidities between subjects with early-onset sporadic ADD (EOSADD; subjects dying under age 60) versus ADD associated with different types of PSEN1 mutations, the most common cause of early-onset autosomal dominant ADD. In particular, we were able to ascertain, for the first time, the prevalences of a fairly complete set of ADD comorbidities in United States (US) PSEN1 cases as well as the Colombian E280A PSEN1 kindred. Data for EOSADD and US PSEN1 subjects (with multiple different mutation types) was obtained from the National Alzheimer Coordinating Center (NACC). Colombian cases all had the E280A mutation and had a set of neuropathological observations classified, like the US cases according to the NACC NP10 definitions. Confirmatory of earlier reports, NACC-defined Alzheimer Disease Neuropathological Changes (ADNC) were consistently very severe in early-onset cases, whether sporadic or in PSEN1 cases, but were slightly less severe in EOSADD. Amyloid angiopathy was the only AD-associated pathology type with widely-differing severity scores between the 3 groups, with median scores of 3, 2 and 1 in the PSEN1 Colombia, PSEN1 US and EOSADD cases, respectively. Apoliprotein E genotype did not show significant proportional group differences for the possession of an E-4 or E-2 allele. Of ADD comorbidities, LBD was most common, being present in more than half of all cases in all 3 groups. For TDP-43 co-pathology, the Colombian PSEN1 group was the most affected, at about 27%, vs 16% and 11% for the US PSEN1 and sporadic US cases, respectively. Notably, hippocampal sclerosis and non-AD tau pathological conditions were not present in any of the US or Colombian PSEN1 cases, and was seen in only 3% of the EOSADD cases. Significant large-vessel atherosclerosis was present in a much larger percentage of Colombian PSEN1 cases, at almost 20% as compared to 0% and 3% of the US PSEN1 and EOSADD cases, respectively. Small-vessel disease, or arteriolosclerosis, was much more common than large vessel disease, being present in all groups between 18% and 37%. Gross and microscopic infarcts, however, as well as gross or microscopic hemorrhages, were generally absent or present at very low percentages in all groups. White matter rarefaction (WMR) was remarkably common, at almost 60%, in the US PSEN1 group, as compared to about 18% in the EOSADD cases, a significant difference. White matter rarefaction was not assessed in the Colombian PSEN1 cases. The results presented here, as well as other evidence, indicates that LBD, TDP-43 pathology and WMR, as common comorbidities with autosomal dominant and early-onset sporadic ADD, should be considered when planning clinical trials with such subjects as they may increase variability in response rates. However, they may be at least partially dependent on ADNC and thus potentially addressable by anti-amyloid or and/anti-tau therapies.
Collapse
Affiliation(s)
- Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52 20246 Hamburg, Gebäude Nord 27 / Raum 02.005
| | | | - Charles White
- Neuropathology Section, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, 10515 W Santa Fe Drive, Sun City, AZ 85351
| | - Juliana Acosta-Uribe
- Faculty of Medicine, Neuroscience Group of Antioquia, University of Antioquia, Medellin, Colombia
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara
| | - Barbara Mejía-Cupajita
- Faculty of Medicine, Neuroscience Group of Antioquia, University of Antioquia, Medellin, Colombia
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara
| | - Nelson David Villalba-Moreno
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52 20246 Hamburg, Gebäude Nord 27 / Raum 02.005
| | - Pinzhang Lu
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52 20246 Hamburg, Gebäude Nord 27 / Raum 02.005
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Martinistraße 52 20246 Hamburg, Gebäude Nord 27 / Raum 02.005
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Matthew P Frosch
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, 10515 W Santa Fe Drive, Sun City, AZ 85351
| |
Collapse
|
36
|
Iadecola C, Anfray A, Schaeffer S, Hattori Y, Santisteban M, Casey N, Wang G, Strickland M, Zhou P, Holtzman D, Anrather J, Park L. Cell autonomous role of border associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. RESEARCH SQUARE 2023:rs.3.rs-3222611. [PMID: 37577565 PMCID: PMC10418550 DOI: 10.21203/rs.3.rs-3222611/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Apolipoprotein-E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4 we report that border associated macrophages (BAM), myeloid cells closely apposed to neocortical microvessels, are both the source and the target of the ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAM is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Wang
- Feil Family Brain and Mind Research Institute - Weill Cornell Medicine
| | | | | | | | | | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
37
|
Ichimata S, Martinez-Valbuena I, Lee S, Li J, Karakani AM, Kovacs GG. Distinct Molecular Signatures of Amyloid-Beta and Tau in Alzheimer's Disease Associated with Down Syndrome. Int J Mol Sci 2023; 24:11596. [PMID: 37511361 PMCID: PMC10380583 DOI: 10.3390/ijms241411596] [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: 06/19/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Limited comparative data exist on the molecular spectrum of amyloid-beta (Aβ) and tau deposition in individuals with Down syndrome (DS) and sporadic Alzheimer's disease (sAD). We assessed Aβ and tau deposition severity in the temporal lobe and cerebellum of ten DS and ten sAD cases. Immunohistochemistry was performed using antibodies against eight different Aβ epitopes (6F/3D, Aβ38, Aβ39, Aβ40, Aβ42, Aβ43, pyroglutamate Aβ at third glutamic acid (AβNp3E), phosphorylated- (p-)Aβ at 8th serine (AβpSer8)), and six different pathological tau epitopes (p-Ser202/Thr205, p-Thr231, p-Ser396, Alz50, MC1, GT38). Findings were evaluated semi-quantitatively and quantitatively using digital pathology. DS cases had significantly higher neocortical parenchymal deposition (Aβ38, Aβ42, and AβpSer8), and cerebellar parenchymal deposition (Aβ40, Aβ42, AβNp3E, and AβpSer8) than sAD cases. Furthermore, DS cases had a significantly larger mean plaque size (6F/3D, Aβ42, AβNp3E) in the temporal lobe, and significantly greater deposition of cerebral and cerebellar Aβ42 than sAD cases in the quantitative analysis. Western blotting corroborated these findings. Regarding tau pathology, DS cases had significantly more severe cerebral tau deposition than sAD cases, especially in the white matter (p-Ser202/Thr205, p-Thr231, Alz50, and MC1). Greater total tau deposition in the white matter (p-Ser202/Thr205, p-Thr231, and Alz50) of DS cases was confirmed by quantitative analysis. Our data suggest that the Aβ and tau molecular signatures in DS are distinct from those in sAD.
Collapse
Affiliation(s)
- Shojiro Ichimata
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Legal Medicine, Faculty of Medicine, University of Toyama, Toyama 930-8555, Japan
| | - Ivan Martinez-Valbuena
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Seojin Lee
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Jun Li
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Ali M. Karakani
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON M5T 2S8, Canada; (S.I.); (I.M.-V.); (S.L.); (J.L.); (A.M.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Edmond J. Safra Program in Parkinson’s Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada
- Laboratory Medicine Program, Krembil Brain Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
38
|
Luo J, Ma Y, Agboola FJ, Grant E, Morris JC, McDade E, Fagan AM, Benzinger TLS, Hassenstab J, Bateman RJ, Perrin RJ, Gordon BA, Goyal M, Strain JF, Yakushev I, Day GS, Xiong C. Longitudinal Relationships of White Matter Hyperintensities and Alzheimer Disease Biomarkers Across the Adult Life Span. Neurology 2023; 101:e164-e177. [PMID: 37202169 PMCID: PMC10351551 DOI: 10.1212/wnl.0000000000207378] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 03/20/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND AND OBJECTIVES White matter hyperintensities (WMH) correlate with Alzheimer disease (AD) biomarkers cross-sectionally and modulate AD pathogenesis. Longitudinal changes have been reported for AD biomarkers, including concentrations of CSF β-amyloid (Aβ) 42, Aβ40, total tau and phosphorylated tau181, standardized uptake value ratio from the molecular imaging of cerebral fibrillar Aβ with PET using [11C] Pittsburgh Compound-B, MRI-based hippocampal volume, and cortical thickness. Correlations between established AD biomarkers and the longitudinal change for WMH have not been fully evaluated, especially among cognitively normal individuals across the adult life span. METHODS We jointly analyzed the longitudinal data of WMH volume and each of the established AD biomarkers and cognition from 371 cognitively normal individuals whose baseline age spanned from 19.6 to 88.20 years from 4 longitudinal studies of aging and AD. A 2-stage algorithm was applied to identify the inflection point of baseline age whereby older participants had an accelerated longitudinal change in WMH volume, in comparison with the younger participants. The longitudinal correlations between WMH volume and AD biomarkers were estimated from the bivariate linear mixed-effects models. RESULTS A longitudinal increase in WMH volume was associated with a longitudinal increase in PET amyloid uptake and a decrease in MRI hippocampal volume, cortical thickness, and cognition. The inflection point of baseline age in WMH volume was identified at 60.46 (95% CI 56.43-64.49) years, with the annual increase for the older participants (83.12 [SE = 10.19] mm3 per year) more than 13 times faster (p < 0.0001) than that for the younger participants (6.35 [SE = 5.63] mm3 per year). Accelerated rates of change among the older participants were similarly observed in almost all the AD biomarkers. Longitudinal correlations of WMH volume with MRI, PET amyloid biomarkers, and cognition seemed to be numerically stronger for the younger participants, but not significantly different from those for the older participants. Carrying APOE ε4 alleles did not alter the longitudinal correlations between WMH and AD biomarkers. DISCUSSION Longitudinal increases in WMH volume started to accelerate around a baseline age of 60.46 years and correlated with the longitudinal change in PET amyloid uptake, MRI structural outcomes, and cognition.
Collapse
Affiliation(s)
- Jingqin Luo
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Yinjiao Ma
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Folasade Jane Agboola
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Elizabeth Grant
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - John C Morris
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Eric McDade
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Anne M Fagan
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Tammie L S Benzinger
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Jason Hassenstab
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Randall J Bateman
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Richard J Perrin
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Brian A Gordon
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Manu Goyal
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Jeremy F Strain
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Igor Yakushev
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Gregory S Day
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Chengjie Xiong
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
39
|
Ruan Z, Sun D, Zhou X, Yu M, Li S, Sun W, Li Y, Gao L, Xu H. Altered neurovascular coupling in patients with vascular cognitive impairment: a combined ASL-fMRI analysis. Front Aging Neurosci 2023; 15:1224525. [PMID: 37416325 PMCID: PMC10320594 DOI: 10.3389/fnagi.2023.1224525] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 06/08/2023] [Indexed: 07/08/2023] Open
Abstract
Background and objective This study aims to examine the role of neurovascular coupling (NVC) in vascular cognitive impairment (VCI) by investigating the relationship between white matter lesion (WML) burden, NVC, and cognitive deficits. Additionally, we aim to explore the potential of NVC as a tool for understanding the neural mechanisms underlying VCI. Methods This study included thirty-eight small vessel disease cognitive impairment (SVCI) patients, 34 post-stroke cognitive impairment (PSCI) patients, and 43 healthy controls (HC). Comprehensive assessments, including neuroimaging and neuropsychological testing, were conducted to evaluate cognitive function. WML burden was measured and correlated with NVC coefficients to examine the relationship between white matter pathology and NVC. Mediation analysis was employed to explore the link relationship between NVC, WML burden, and cognitive function. Results The present study showed that NVC was significantly reduced in the SVCI and PSCI groups compared with HCs at both whole-brain and brain region level. The analysis revealed notable findings regarding NVC in relation to WML burden and cognitive function in VCI patients. Specifically, reduced NVC coefficients were observed within higher order brain systems responsible for cognitive control and emotion regulation. Mediation analysis demonstrated that NVC played a mediating role in the relationship between WML burden and cognitive impairment. Conclusion This study reveals the mediating role of NVC in the relationship between WML burden and cognitive function in VCI patients. The results demonstrate the potential of the NVC as an accurate measure of cognitive impairment and its ability to identify specific neural circuits affected by WML burden.
Collapse
Affiliation(s)
- Zhao Ruan
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Dong Sun
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaoli Zhou
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Minhua Yu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenbo Sun
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yidan Li
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lei Gao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
40
|
Pansieri J, Hadley G, Lockhart A, Pisa M, DeLuca GC. Regional contribution of vascular dysfunction in white matter dementia: clinical and neuropathological insights. Front Neurol 2023; 14:1199491. [PMID: 37396778 PMCID: PMC10313211 DOI: 10.3389/fneur.2023.1199491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
The maintenance of adequate blood supply and vascular integrity is fundamental to ensure cerebral function. A wide range of studies report vascular dysfunction in white matter dementias, a group of cerebral disorders characterized by substantial white matter damage in the brain leading to cognitive impairment. Despite recent advances in imaging, the contribution of vascular-specific regional alterations in white matter dementia has been not extensively reviewed. First, we present an overview of the main components of the vascular system involved in the maintenance of brain function, modulation of cerebral blood flow and integrity of the blood-brain barrier in the healthy brain and during aging. Second, we review the regional contribution of cerebral blood flow and blood-brain barrier disturbances in the pathogenesis of three distinct conditions: the archetypal white matter predominant neurocognitive dementia that is vascular dementia, a neuroinflammatory predominant disease (multiple sclerosis) and a neurodegenerative predominant disease (Alzheimer's). Finally, we then examine the shared landscape of vascular dysfunction in white matter dementia. By emphasizing the involvement of vascular dysfunction in the white matter, we put forward a hypothetical map of vascular dysfunction during disease-specific progression to guide future research aimed to improve diagnostics and facilitate the development of tailored therapies.
Collapse
|
41
|
Cogswell PM, Lundt ES, Therneau TM, Mester CT, Wiste HJ, Graff-Radford J, Schwarz CG, Senjem ML, Gunter JL, Reid RI, Przybelski SA, Knopman DS, Vemuri P, Petersen RC, Jack CR. Evidence against a temporal association between cerebrovascular disease and Alzheimer's disease imaging biomarkers. Nat Commun 2023; 14:3097. [PMID: 37248223 PMCID: PMC10226977 DOI: 10.1038/s41467-023-38878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 05/15/2023] [Indexed: 05/31/2023] Open
Abstract
Whether a relationship exists between cerebrovascular disease and Alzheimer's disease has been a source of controversy. Evaluation of the temporal progression of imaging biomarkers of these disease processes may inform mechanistic associations. We investigate the relationship of disease trajectories of cerebrovascular disease (white matter hyperintensity, WMH, and fractional anisotropy, FA) and Alzheimer's disease (amyloid and tau PET) biomarkers in 2406 Mayo Clinic Study of Aging and Mayo Alzheimer's Disease Research Center participants using accelerated failure time models. The model assumes a common pattern of progression for each biomarker that is shifted earlier or later in time for each individual and represented by a per participant age adjustment. An individual's amyloid and tau PET adjustments show very weak temporal association with WMH and FA adjustments (R = -0.07 to 0.07); early/late amyloid or tau timing explains <1% of the variation in WMH and FA adjustment. Earlier onset of amyloid is associated with earlier onset of tau (R = 0.57, R2 = 32%). These findings support a strong mechanistic relationship between amyloid and tau aggregation, but not between WMH or FA and amyloid or tau PET.
Collapse
Affiliation(s)
- Petrice M Cogswell
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| | - Emily S Lundt
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Terry M Therneau
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Carly T Mester
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Heather J Wiste
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | | | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Department of Information Technology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Jeffrey L Gunter
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Robert I Reid
- Department of Information Technology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Ronald C Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Department of Neurology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| |
Collapse
|
42
|
Beach TG, Sue LI, Scott S, Intorcia AJ, Walker JE, Arce RA, Glass MJ, Borja CI, Cline MP, Hemmingsen SJ, Qiji S, Stewart A, Martinez KN, Krupp A, McHattie R, Mariner M, Lorenzini I, Kuramoto A, Long KE, Tremblay C, Caselli RJ, Woodruff BK, Rapscak SZ, Belden CM, Goldfarb D, Choudhury P, Driver-Dunckley ED, Mehta SH, Sabbagh MN, Shill HA, Atri A, Adler CH, Serrano GE. Cerebral white matter rarefaction has both neurodegenerative and vascular causes and may primarily be a distal axonopathy. J Neuropathol Exp Neurol 2023; 82:457-466. [PMID: 37071794 PMCID: PMC10209646 DOI: 10.1093/jnen/nlad026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
Cerebral white matter rarefaction (CWMR) was considered by Binswanger and Alzheimer to be due to cerebral arteriolosclerosis. Renewed attention came with CT and MR brain imaging, and neuropathological studies finding a high rate of CWMR in Alzheimer disease (AD). The relative contributions of cerebrovascular disease and AD to CWMR are still uncertain. In 1181 autopsies by the Arizona Study of Aging and Neurodegenerative Disorders (AZSAND), large-format brain sections were used to grade CWMR and determine its vascular and neurodegenerative correlates. Almost all neurodegenerative diseases had more severe CWMR than the normal control group. Multivariable logistic regression models indicated that Braak neurofibrillary stage was the strongest predictor of CWMR, with additional independently significant predictors including age, cortical and diencephalic lacunar and microinfarcts, body mass index, and female sex. It appears that while AD and cerebrovascular pathology may be additive in causing CWMR, both may be solely capable of this. The typical periventricular pattern suggests that CWMR is primarily a distal axonopathy caused by dysfunction of the cell bodies of long-association corticocortical projection neurons. A consequence of these findings is that CWMR should not be viewed simply as "small vessel disease" or as a pathognomonic indicator of vascular cognitive impairment or vascular dementia.
Collapse
Affiliation(s)
- Thomas G Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Sarah Scott
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | - Richard A Arce
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Michael J Glass
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Madison P Cline
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Sanaria Qiji
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Analisa Stewart
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Addison Krupp
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Rylee McHattie
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Monica Mariner
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | - Angela Kuramoto
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Kathy E Long
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | | | | | | | | | | | | | | - Shyamal H Mehta
- Department of Neurology, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Holly A Shill
- Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, Arizona, USA
- Harvard Medical School & Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Charles H Adler
- Department of Neurology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Geidy E Serrano
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
43
|
Kamal F, Morrison C, Dadar M. Investigating the relationship between sleep disturbances and white matter hyperintensities in older adults on the Alzheimer's disease spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.13.23288544. [PMID: 37131746 PMCID: PMC10153314 DOI: 10.1101/2023.04.13.23288544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background While studies report that sleep disturbance can have negative effects on brain vasculature, its impact on cerebrovascular disease such as white matter hyperintensities (WMHs) in beta-amyloid positive older adults remains unexplored. Methods Linear regressions, mixed effects models, and mediation analysis examined the crosssectional and longitudinal associations between sleep disturbance, cognition, and WMH burden, and cognition in normal controls (NCs), mild cognitive impairment (MCI), and Alzheimer's disease (AD) at baseline and longitudinally. Results People with AD reported more sleep disturbance than NC and MCI. AD with sleep disturbance had more WMHs than AD without sleep disturbances. Mediation analysis revealed an effect of regional WMH burden on the relationship between sleep disturbance and future cognition. Conclusion These results suggest that WMH burden and sleep disturbance increases from aging to AD. Sleep disturbance decreases cognition through increases in WMH burden. Improved sleep could mitigate the impact of WMH accumulation and cognitive decline.
Collapse
Affiliation(s)
- Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Cassandra Morrison
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| |
Collapse
|
44
|
Chan K, Fischer C, Maralani PJ, Black SE, Moody AR, Khademi A. Alzheimer's and vascular disease classification using regional texture biomarkers in FLAIR MRI. Neuroimage Clin 2023; 38:103385. [PMID: 36989851 PMCID: PMC10074987 DOI: 10.1016/j.nicl.2023.103385] [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: 12/03/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Interactions between subcortical vascular disease and dementia due to Alzheimer's disease (AD) are unclear, and clinical overlap between the diseases makes diagnosis challenging. Existing studies have shown regional microstructural changes specific to each disease, and that textures in fluid-attenuated inversion recovery (FLAIR) MRI images may characterize abnormalities in tissue microstructure. This work aims to investigate regional FLAIR biomarkers that can differentiate dementia cohorts with and without subcortical vascular disease. FLAIR and diffusion MRI (dMRI) volumes were obtained in 65 mild cognitive impairment (MCI), 21 AD, 44 subcortical vascular MCI (scVMCI), 22 Mixed etiology, and 48 healthy elderly patients. FLAIR texture and intensity biomarkers were extracted from the normal appearing brain matter (NABM), WML penumbra, blood supply territory (BST), and white matter tract regions of each patient. All FLAIR biomarkers were correlated to dMRI metrics in each region and global WML load, and biomarker means between groups were compared using ANOVA. Binary classifications were performed using Random Forest classifiers to investigate the predictive nature of the regional biomarkers, and SHAP feature analysis was performed to further investigate optimal regions of interest for differentiating disease groups. The regional FLAIR biomarkers were strongly correlated to MD, while all biomarker regions but white matter tracts were strongly correlated to WML burden. Classification between Mixed disease and healthy, AD, and scVMCI patients yielded accuracies of 97%, 81%, and 72% respectively using WM tract biomarkers. Classification between scVMCI and healthy, MCI, and AD patients yielded accuracies of 89%, 84%, and 79% respectively using penumbra biomarkers. Only the classification between AD and healthy patients had optimal results using NABM biomarkers. This work presents novel regional FLAIR biomarkers that may quantify white matter degeneration related to subcortical vascular disease, and which indicate that investigating degeneration in specific regions may be more important than assessing global WML burden in vascular disease groups.
Collapse
Affiliation(s)
- Karissa Chan
- Electrical, Computer and Biomedical Engineering Department, Toronto Metropolitan University, 350 Victoria St., Toronto, ON M5B 2K3, Canada; Institute for Biomedical Engineering, Science Tech (iBEST), A Partnership Between St. Michael's Hospital and Toronto Metropolitan University, 209 Victoria St., Toronto, ON M5B 1T8, Canada.
| | - Corinne Fischer
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Network, 30 Bond St., Toronto, ON M5B 1W8, Canada; Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada.
| | - Pejman Jabehdar Maralani
- Department of Medical Imaging, University of Toronto, 263 McCaul St., Toronto, ON M5T 1W7, Canada.
| | - Sandra E Black
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Horvitz Brain Sciences Research Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada.
| | - Alan R Moody
- Department of Medical Imaging, University of Toronto, 263 McCaul St., Toronto, ON M5T 1W7, Canada.
| | - April Khademi
- Electrical, Computer and Biomedical Engineering Department, Toronto Metropolitan University, 350 Victoria St., Toronto, ON M5B 2K3, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Network, 30 Bond St., Toronto, ON M5B 1W8, Canada; Institute for Biomedical Engineering, Science Tech (iBEST), A Partnership Between St. Michael's Hospital and Toronto Metropolitan University, 209 Victoria St., Toronto, ON M5B 1T8, Canada; Rotman Research Institute, Baycrest Hospital, 3560 Bathurst Street, Toronto, ON M6A 2E1, Canada.
| |
Collapse
|
45
|
Berk-Rauch HE, Choudhury A, Richards AT, Singh PK, Chen ZL, Norris EH, Strickland S, Ahn HJ. Striatal fibrinogen extravasation and vascular degeneration correlate with motor dysfunction in an aging mouse model of Alzheimer’s disease. Front Aging Neurosci 2023; 15:1064178. [PMID: 36967821 PMCID: PMC10034037 DOI: 10.3389/fnagi.2023.1064178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Alzheimer’s Disease (AD) patients exhibit signs of motor dysfunction, including gait, locomotion, and balance deficits. Changes in motor function often precede other symptoms of AD as well as correlate with increased severity and mortality. Despite the frequent occurrence of motor dysfunction in AD patients, little is known about the mechanisms by which this behavior is altered.Methods and Results: In the present study, we investigated the relationship between cerebrovascular impairment and motor dysfunction in a mouse model of AD (Tg6799). We found an age-dependent increase of extravasated fibrinogen deposits in the cortex and striatum of AD mice. Interestingly, there was significantly decreased cerebrovascular density in the striatum of the 15-month-old as compared to 7-month-old AD mice. We also found significant demyelination and axonal damage in the striatum of aged AD mice. We analyzed striatum-related motor function and anxiety levels of AD mice at both ages and found that aged AD mice exhibited significant impairment of motor function but not in the younger AD mice.Discussion: Our finding suggests an enticing correlation between extravasated fibrinogen, cerebrovascular damage of the striatum, and motor dysfunction in an AD mouse model, suggesting a possible mechanism underlying motor dysfunction in AD.
Collapse
Affiliation(s)
- Hanna E. Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Arnab Choudhury
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Allison T. Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Pradeep K. Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, United States
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- *Correspondence: Hyung Jin Ahn,
| |
Collapse
|
46
|
Alosco ML, Ly M, Mosaheb S, Saltiel N, Uretsky M, Tripodis Y, Martin B, Palmisano J, Delano-Wood L, Bondi MW, Meng G, Xia W, Daley S, Goldstein LE, Katz DI, Dwyer B, Daneshvar DH, Nowinski C, Cantu RC, Kowall NW, Stern RA, Alvarez VE, Mez J, Huber BR, McKee AC, Stein TD. Decreased myelin proteins in brain donors exposed to football-related repetitive head impacts. Brain Commun 2023; 5:fcad019. [PMID: 36895961 PMCID: PMC9990992 DOI: 10.1093/braincomms/fcad019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/02/2022] [Accepted: 01/30/2023] [Indexed: 03/09/2023] Open
Abstract
American football players and other individuals exposed to repetitive head impacts can exhibit a constellation of later-life cognitive and neuropsychiatric symptoms. While tau-based diseases such as chronic traumatic encephalopathy can underpin certain symptoms, contributions from non-tau pathologies from repetitive head impacts are increasingly recognized. We examined cross-sectional associations between myelin integrity using immunoassays for myelin-associated glycoprotein and proteolipid protein 1 with risk factors and clinical outcomes in brain donors exposed to repetitive head impacts from American football. Immunoassays for myelin-associated glycoprotein and proteolipid protein 1 were conducted on dorsolateral frontal white matter tissue samples of 205 male brain donors. Proxies of exposure to repetitive head impacts included years of exposure and age of first exposure to American football play. Informants completed the Functional Activities Questionnaire, Behavior Rating Inventory of Executive Function-Adult Version (Behavioral Regulation Index), and Barratt Impulsiveness Scale-11. Associations between myelin-associated glycoprotein and proteolipid protein 1 with exposure proxies and clinical scales were tested. Of the 205 male brain donors who played amateur and professional football, the mean age was 67.17 (SD = 16.78), and 75.9% (n = 126) were reported by informants to be functionally impaired prior to death. Myelin-associated glycoprotein and proteolipid protein 1 correlated with the ischaemic injury scale score, a global indicator of cerebrovascular disease (r = -0.23 and -0.20, respectively, Ps < 0.01). Chronic traumatic encephalopathy was the most common neurodegenerative disease (n = 151, 73.7%). Myelin-associated glycoprotein and proteolipid protein 1 were not associated with chronic traumatic encephalopathy status, but lower proteolipid protein 1 was associated with more severe chronic traumatic encephalopathy (P = 0.03). Myelin-associated glycoprotein and proteolipid protein 1 were not associated with other neurodegenerative disease pathologies. More years of football play was associated with lower proteolipid protein 1 [beta = -2.45, 95% confidence interval (CI) [-4.52, -0.38]] and compared with those who played <11 years of football (n = 78), those who played 11 or more years (n = 128) had lower myelin-associated glycoprotein (mean difference = 46.00, 95% CI [5.32, 86.69]) and proteolipid protein 1 (mean difference = 24.72, 95% CI [2.40, 47.05]). Younger age of first exposure corresponded to lower proteolipid protein 1 (beta = 4.35, 95% CI [0.25, 8.45]). Among brain donors who were aged 50 or older (n = 144), lower proteolipid protein 1 (beta = -0.02, 95% CI [-0.047, -0.001]) and myelin-associated glycoprotein (beta = -0.01, 95% CI [-0.03, -0.002]) were associated with higher Functional Activities Questionnaire scores. Lower myelin-associated glycoprotein correlated with higher Barratt Impulsiveness Scale-11 scores (beta = -0.02, 95% CI [-0.04, -0.0003]). Results suggest that decreased myelin may represent a late effect of repetitive head impacts that contributes to the manifestation of cognitive symptoms and impulsivity. Clinical-pathological correlation studies with prospective objective clinical assessments are needed to confirm our findings.
Collapse
Affiliation(s)
- Michael L Alosco
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Monica Ly
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
| | - Sydney Mosaheb
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Nicole Saltiel
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Madeline Uretsky
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Brett Martin
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Joseph Palmisano
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Lisa Delano-Wood
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
| | - Mark W Bondi
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego Health, La Jolla, CA, USA
| | | | - Weiming Xia
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sarah Daley
- VA Bedford Healthcare System, Bedford, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lee E Goldstein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Biomedical, Electrical & Computer Engineering, Boston University College of Engineering, Boston, MA, USA
| | - Douglas I Katz
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Brigid Dwyer
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Daniel H Daneshvar
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Robert C Cantu
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Concussion Legacy Foundation, Boston, MA, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Emerson Hospital, Concord, MA, USA
| | - Neil W Kowall
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, Boston, MA, USA
| | - Robert A Stern
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Victor E Alvarez
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, Boston, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, Boston, MA, USA
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Bertrand Russell Huber
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, Boston, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, Boston, MA, USA
| | - Ann C McKee
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Departments of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, Boston, MA, USA
- National Center for PTSD, VA Boston Healthcare, Jamaica Plain, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer’s Disease Research Center and CTE Center, Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Bedford Healthcare System, Bedford, MA, USA
- Departments of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- VA Boston Healthcare System, U.S. Department of Veteran Affairs, Jamaica Plain, Boston, MA, USA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
47
|
Rashid T, Li K, Toledo JB, Nasrallah I, Pajewski NM, Dolui S, Detre J, Wolk DA, Liu H, Heckbert SR, Bryan RN, Williamson J, Davatzikos C, Seshadri S, Launer LJ, Habes M. Association of Intensive vs Standard Blood Pressure Control With Regional Changes in Cerebral Small Vessel Disease Biomarkers: Post Hoc Secondary Analysis of the SPRINT MIND Randomized Clinical Trial. JAMA Netw Open 2023; 6:e231055. [PMID: 36857053 PMCID: PMC9978954 DOI: 10.1001/jamanetworkopen.2023.1055] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
IMPORTANCE Little is known about the associations of strict blood pressure (BP) control with microstructural changes in small vessel disease markers. OBJECTIVE To investigate the regional associations of intensive vs standard BP control with small vessel disease biomarkers, such as white matter lesions (WMLs), fractional anisotropy (FA), mean diffusivity (MD), and cerebral blood flow (CBF). DESIGN, SETTING, AND PARTICIPANTS The Systolic Blood Pressure Intervention Trial (SPRINT) is a multicenter randomized clinical trial that compared intensive systolic BP (SBP) control (SBP target <120 mm Hg) vs standard control (SBP target <140 mm Hg) among participants aged 50 years or older with hypertension and without diabetes or a history of stroke. The study began randomization on November 8, 2010, and stopped July 1, 2016, with a follow-up duration of approximately 4 years. A total of 670 and 458 participants completed brain magnetic resonance imaging at baseline and follow-up, respectively, and comprise the cohort for this post hoc analysis. Statistical analyses for this post hoc analysis were performed between August 2020 and October 2022. INTERVENTIONS At baseline, 355 participants received intensive SBP treatment and 315 participants received standard SBP treatment. MAIN OUTCOMES AND MEASURES The main outcomes were regional changes in WMLs, FA, MD (in white matter regions of interest), and CBF (in gray matter regions of interest). RESULTS At baseline, 355 participants (mean [SD] age, 67.7 [8.0] years; 200 men [56.3%]) received intensive BP treatment and 315 participants (mean [SD] age, 67.0 [8.4] years; 199 men [63.2%]) received standard BP treatment. Intensive treatment was associated with smaller mean increases in WML volume compared with standard treatment (644.5 mm3 vs 1258.1 mm3). The smaller mean increases were observed specifically in the deep white matter regions of the left anterior corona radiata (intensive treatment, 30.3 mm3 [95% CI, 16.0-44.5 mm3]; standard treatment, 80.5 mm3 [95% CI, 53.8-107.2 mm3]), left tapetum (intensive treatment, 11.8 mm3 [95% CI, 4.4-19.2 mm3]; standard treatment, 27.2 mm3 [95% CI, 19.4-35.0 mm3]), left superior fronto-occipital fasciculus (intensive treatment, 3.2 mm3 [95% CI, 0.7-5.8 mm3]; standard treatment, 9.4 mm3 [95% CI, 5.5-13.4 mm3]), left posterior corona radiata (intensive treatment, 26.0 mm3 [95% CI, 12.9-39.1 mm3]; standard treatment, 52.3 mm3 [95% CI, 34.8-69.8 mm3]), left splenium of the corpus callosum (intensive treatment, 45.4 mm3 [95% CI, 25.1-65.7 mm3]; standard treatment, 83.0 mm3 [95% CI, 58.7-107.2 mm3]), left posterior thalamic radiation (intensive treatment, 53.0 mm3 [95% CI, 29.8-76.2 mm3]; standard treatment, 106.9 mm3 [95% CI, 73.4-140.3 mm3]), and right posterior thalamic radiation (intensive treatment, 49.5 mm3 [95% CI, 24.3-74.7 mm3]; standard treatment, 102.6 mm3 [95% CI, 71.0-134.2 mm3]). CONCLUSIONS AND RELEVANCE This study suggests that intensive BP treatment, compared with standard treatment, was associated with a slower increase of WMLs, improved diffusion tensor imaging, and FA and CBF changes in several brain regions that represent vulnerable areas that may benefit from more strict BP control. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01206062.
Collapse
Affiliation(s)
- Tanweer Rashid
- 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
| | - Karl Li
- 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
| | - Jon B. Toledo
- Department of Neurology, University of Florida, Gainesville
- Department of Neurology, Houston Methodist Hospital, Houston, Texas
| | - Ilya Nasrallah
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Nicholas M. Pajewski
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sudipto Dolui
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - John Detre
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Hangfan Liu
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | | | - R. Nick Bryan
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| | - Jeff Williamson
- Section of Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Christos Davatzikos
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Sudha Seshadri
- 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
| | - Lenore J. Launer
- Intramural Research Program, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Mohamad Habes
- 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
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia
| |
Collapse
|
48
|
Morrison C, Dadar M, Manera AL, Collins DL. Racial differences in white matter hyperintensity burden in older adults. Neurobiol Aging 2023; 122:112-119. [PMID: 36543016 DOI: 10.1016/j.neurobiolaging.2022.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
White matter hyperintensities (WMHs) may be one of the earliest pathological changes in aging. Race differences in WMH burden has been conflicting. This study examined if race influences WMHs and whether these differences are influenced by vascular risk factors. Alzheimer's Disease Neuroimaging Initiative participants were included if they had a baseline MRI, diagnosis, and WMH measurements. Ninety-one Blacks and 1937 Whites were included. Using bootstrap re-sampling, 91 Whites were randomly sampled and matched to Blacks based on age, sex, education, and diagnosis 1000 times. Linear models examined the influence of race on baseline WMHs, and change of WMHs over time, with and without vascular factors. Vascular risk factors had higher prevalence in Blacks than Whites. When not including vascular factors, Blacks had greater frontal, parietal, deep, and total WMH burden compared to Whites. There were no race differences in longitudinal progression of WMH accumulation. After controlling for vascular factors, only overall longitudinal parietal WMH group differences remained significant, suggesting that vascular factors contribute to racial group differences observed in WMHs.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Ana L Manera
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Morrison C, Dadar M, Villeneuve S, Ducharme S, Collins DL. White matter hyperintensity load varies depending on subjective cognitive decline criteria. GeroScience 2023; 45:17-28. [PMID: 36401741 PMCID: PMC9886741 DOI: 10.1007/s11357-022-00684-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022] Open
Abstract
Increased age and cognitive impairment is associated with an increase in cerebrovascular pathology often measured as white matter hyperintensities (WMHs) on MRI. Whether WMH burden differs between cognitively unimpaired older adults with subjective cognitive decline (SCD +) and without subjective cognitive decline (SCD -) remains conflicting, and could be related to the methods used to identify SCD. Our goal was to examine if four common SCD classification methods are associated with different WMH accumulation patterns between SCD + and SCD - . A total of 535 cognitively unimpaired older adults with 1353 time points from the Alzheimer's Disease Neuroimaging Initiative were included in this study. SCD was operationalized using four different methods: Cognitive Change Index (CCI), Everyday Cognition Scale (ECog), ECog + Worry, and Worry. Linear mixed-effects models were used to investigate the associations between SCD and overall and regional WMH burden. Overall temporal WMH burden differences were only observed with the Worry questionnaire. Higher WMH burden change over time was observed in SCD + compared to SCD - in the temporal and parietal regions using the CCI (temporal, p = .01; parietal p = .02) and ECog (temporal, p = .02; parietal p = .01). For both the ECog + Worry and Worry questionnaire, change in WMH burden over time was increased in SCD + compared to SCD - for overall, frontal, temporal, and parietal WMH burden (p < .05). These results show that WMH burden differs between SCD + and SCD - depending on the questionnaire and the approach (regional/global) used to measure WMHs. The various methods used to define SCD may reflect different types of underlying pathologies.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
50
|
Toledo JB, Abdelnour C, Weil RS, Ferreira D, Rodriguez-Porcel F, Pilotto A, Wyman-Chick KA, Grothe MJ, Kane JPM, Taylor A, Rongve A, Scholz S, Leverenz JB, Boeve BF, Aarsland D, McKeith IG, Lewis S, Leroi I, Taylor JP. Dementia with Lewy bodies: Impact of co-pathologies and implications for clinical trial design. Alzheimers Dement 2023; 19:318-332. [PMID: 36239924 PMCID: PMC9881193 DOI: 10.1002/alz.12814] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 02/01/2023]
Abstract
Dementia with Lewy bodies (DLB) is clinically defined by the presence of visual hallucinations, fluctuations, rapid eye movement (REM) sleep behavioral disorder, and parkinsonism. Neuropathologically, it is characterized by the presence of Lewy pathology. However, neuropathological studies have demonstrated the high prevalence of coexistent Alzheimer's disease, TAR DNA-binding protein 43 (TDP-43), and cerebrovascular pathologic cases. Due to their high prevalence and clinical impact on DLB individuals, clinical trials should account for these co-pathologies in their design and selection and the interpretation of biomarkers values and outcomes. Here we discuss the frequency of the different co-pathologies in DLB and their cross-sectional and longitudinal clinical impact. We then evaluate the utility and possible applications of disease-specific and disease-nonspecific biomarkers and how co-pathologies can impact these biomarkers. We propose a framework for integrating multi-modal biomarker fingerprints and step-wise selection and assessment of DLB individuals for clinical trials, monitoring target engagement, and interpreting outcomes in the setting of co-pathologies.
Collapse
Affiliation(s)
- Jon B Toledo
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA
| | - Carla Abdelnour
- Fundació ACE. Barcelona Alzheimer Treatment and Research Center, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Rimona S Weil
- Dementia Research Centre, Wellcome Centre for Human Neuroimaging, Movement Disorders Consortium, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer's Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, University of Brescia, Parkinson's Disease Rehabilitation Centre, FERB ONLUS-S, Isidoro Hospital, Trescore Balneario (BG), Italy
| | - Kathryn A Wyman-Chick
- HealthPartners Center for Memory and Aging and Struthers Parkinson's Center, Saint Paul, Minnesota, USA
| | - Michel J Grothe
- Instituto de Biomedicina de Sevilla (IBiS), Unidad de Trastornos del Movimiento, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Joseph P M Kane
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Angela Taylor
- Lewy Body Dementia Association, Lilburn, Georgia, USA
| | - Arvid Rongve
- Department of Research and Innovation, Institute of Clinical Medicine (K1), Haugesund Hospital, Norway and The University of Bergen, Bergen, Norway
| | - Sonja Scholz
- Department of Neurology, National Institute of Neurological Disorders and Stroke, Neurodegenerative Diseases Research Unit, Johns Hopkins University Medical Center, Baltimore, Maryland, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bradley F Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Ian G McKeith
- Newcastle University Translational and Clinical Research Institute (NUTCRI, Newcastle upon Tyne, UK
| | - Simon Lewis
- ForeFront Parkinson's Disease Research Clinic, School of Medical Sciences, Brain and Mind Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Iracema Leroi
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - John P Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|