1
|
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
|
2
|
Lin TK, Huang CR, Lin KJ, Hsieh YH, Chen SD, Lin YC, Chao AC, Yang DI. Potential Roles of Hypoxia-Inducible Factor-1 in Alzheimer's Disease: Beneficial or Detrimental? Antioxidants (Basel) 2024; 13:1378. [PMID: 39594520 PMCID: PMC11591038 DOI: 10.3390/antiox13111378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The major pathological characteristics of Alzheimer's disease (AD) include senile plaques and neurofibrillary tangles (NFTs), which are mainly composed of aggregated amyloid-beta (Aβ) peptide and hyperphosphorylated tau protein, respectively. The excessive production of reactive oxygen species (ROS) and neuroinflammation are crucial contributing factors to the pathological mechanisms of AD. Hypoxia-inducible factor-1 (HIF-1) is a transcription factor critical for tissue adaption to low-oxygen tension. Growing evidence has suggested HIF-1 as a potential therapeutic target for AD; conversely, other experimental findings indicate that HIF-1 induction contributes to AD pathogenesis. These previous findings thus point to the complex, even contradictory, roles of HIF-1 in AD. In this review, we first introduce the general pathogenic mechanisms of AD as well as the potential pathophysiological roles of HIF-1 in cancer, immunity, and oxidative stress. Based on current experimental evidence in the literature, we then discuss the possible beneficial as well as detrimental mechanisms of HIF-1 in AD; these sections also include the summaries of multiple chemical reagents and proteins that have been shown to exert beneficial effects in AD via either the induction or inhibition of HIF-1.
Collapse
Affiliation(s)
- Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Chi-Ren Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
- College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan
| | - Kai-Jung Lin
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Yi-Heng Hsieh
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan; (T.-K.L.); (C.-R.H.); (S.-D.C.)
| | - Yi-Chun Lin
- Department of Neurology, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - A-Ching Chao
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 807377, Taiwan
- Department of Neurology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Ding-I Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
3
|
Bachmann D, Saake A, Studer S, Buchmann A, Rauen K, Gruber E, Michels L, Nitsch RM, Hock C, Gietl A, Treyer V. Hypertension and cerebral blood flow in the development of Alzheimer's disease. Alzheimers Dement 2024; 20:7729-7744. [PMID: 39254220 PMCID: PMC11567827 DOI: 10.1002/alz.14233] [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: 02/24/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION We investigated the interactive associations between amyloid and hypertension on the entorhinal cortex (EC) tau and atrophy and the role of cerebral blood flow (CBF) as a shared mechanism by which amyloid and hypertension contribute to EC tau and regional white matter hyperintensities (WMHs). METHODS We analyzed data from older adults without dementia participating in the Add-Tau study (NCT02958670, n = 138) or Alzheimer's Disease Neuroimaging Initiative (ADNI) (n = 523) who had available amyloid-positron emission tomography (PET), tau-PET, fluid-attenuated inversion recovery (FLAIR), and T1-weighted magnetic resonance imaging (MRI). A subsample in both cohorts had available arterial spin labeling (ASL) MRI (Add-Tau: n = 78; ADNI: n = 89). RESULTS The detrimental effects of hypertension on AD pathology and EC thickness were more pronounced in the Add-Tau cohort. Increased amyloid burden was associated with decreased occipital gray matter CBF in the ADNI cohort. In both cohorts, lower regional gray matter CBF was associated with higher EC tau and posterior WMH burden. DISCUSSION Reduced cerebral perfusion may be one common mechanism through which hypertension and amyloid are related to increased EC tau and WMH volume. HIGHLIGHTS Hypertension is associated with increased entorhinal cortex (EC) tau, particularly in the presence of amyloid. Decreased cortical cerebral blood flow (CBF) is associated with higher regional white matter hyperintensity volume. Increasing amyloid burden is associated with decreasing CBF in the occipital lobe. MTL CBF and amyloid are synergistically associated with EC tau.
Collapse
Affiliation(s)
- Dario Bachmann
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Health Sciences and TechnologyETH ZürichZurichSwitzerland
| | - Antje Saake
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Sandro Studer
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Andreas Buchmann
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Katrin Rauen
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Geriatric PsychiatryPsychiatric Hospital ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of ZurichZurichSwitzerland
| | - Esmeralda Gruber
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Lars Michels
- Department of NeuroradiologyClinical Neuroscience Center, University Hospital ZurichZurichSwitzerland
| | - Roger M. Nitsch
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- NeurimmuneZurichSwitzerland
| | - Christoph Hock
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- NeurimmuneZurichSwitzerland
| | - Anton Gietl
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Geriatric PsychiatryPsychiatric Hospital ZurichZurichSwitzerland
| | - Valerie Treyer
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Nuclear MedicineUniversity Hospital of Zurich, University of ZurichZurichSwitzerland
| |
Collapse
|
4
|
Sung KL, Kuo MJ, Yang HY, Tsai CF, Sung SF. Poststroke seizures and epilepsy increase the risk of dementia among stroke survivors: A population-based study. Epilepsia 2024; 65:3244-3254. [PMID: 39254353 DOI: 10.1111/epi.18117] [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: 05/04/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024]
Abstract
OBJECTIVE With global aging, the occurrence of stroke and associated outcomes like dementia are on the rise. Seizures and epilepsy are common poststroke complications and have a strong connection to subsequent dementia. This study examines the relationship between poststroke seizures (PSS) or poststroke epilepsy (PSE) and dementia using a national health care database. METHODS We conducted a retrospective study using data from the Taiwan National Health Insurance Research Database from 2009 to 2020. We identified acute stroke patients from 2010 to 2015, excluding those with pre-existing neurological conditions. Based on age, sex, stroke severity level, and the year of index stroke, patients with PSS or PSE were matched to those without. The main outcome was incident dementia. RESULTS This study included 62 968 patients with an average age of 63 years, with males accounting for 62.9%. Of them, 60.3% had ischemic strokes, and 39.7% had hemorrhagic strokes. After an average follow-up period of 5.2 years, dementia developed in 15.9% of patients who had PSS or PSE, as opposed to 8.4% of those without these conditions. A time-dependent Fine and Gray competing risk analysis showed that PSS and PSE were significantly associated with dementia across all stroke types. Subgroup analyses revealed significantly increased risk of dementia across all age groups (<50, 50-64, and ≥65 years), sexes, and various stroke severity levels. The link between PSS or PSE and dementia was particularly pronounced in men, with a less distinct correlation in women. SIGNIFICANCE The risk of incident dementia was higher in patients with PSS or PSE. The potential for therapeutic interventions for seizures and epilepsy to reduce poststroke dementia underscores the importance of seizure screening and treatment in stroke survivors.
Collapse
Affiliation(s)
- Kuan-Lin Sung
- School of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Miao-Jen Kuo
- School of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsin-Yi Yang
- Clinical Data Center, Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Ching-Fang Tsai
- Clinical Data Center, Department of Medical Research, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Sheng-Feng Sung
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Nursing, Fooyin University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Chatterjee A, Lee S, Diaz V, Saloner R, Sanderson-Cimino M, deCarli C, Maillard P, Hinman J, Vossel K, Casaletto KB, Staffaroni AM, Paolillo EW, Kramer JH. Associations of cerebrovascular disease and Alzheimer's disease pathology with cognitive decline: Analysis of the National Alzheimer's Coordinating Center Uniform Data Set. Neurobiol Aging 2024; 142:1-7. [PMID: 39024720 DOI: 10.1016/j.neurobiolaging.2024.06.002] [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: 10/21/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Cerebrovascular disease (CVD) and Alzheimer's disease (AD) often co-occur and may impact specific cognitive domains. This study's goal was to determine effects of CVD and AD burden on cross-sectional and longitudinal executive function (EF) and memory in older adults. Longitudinally followed participants from the National Alzheimer Coordinating Center database (n = 3342) were included. Cognitive outcomes were EF and memory composite scores. Baseline CVD presence was defined by moderate-to-severe white matter hyperintensities or lacunar infarct on MRI. Baseline AD pathology was defined by amyloid positivity via PET or CSF. Linear mixed models examined effects of CVD, AD, and time on cognitive outcomes, controlling for sex, education, baseline age, MoCA score, and total number of study visits. At baseline, CVD associated with lower EF (p < 0.001), while AD associated with lower EF and memory (ps < 0.001). Longitudinally only AD associated with faster declines in memory and EF (ps < 0.001). These results extend our understanding of CVD and AD pathology, highlighting that CVD does not necessarily indicate accelerated decline.
Collapse
Affiliation(s)
- Ankita Chatterjee
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA.
| | - Shannon Lee
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Valentina Diaz
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Mark Sanderson-Cimino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Charles deCarli
- Department of Neurology, University of California, Davis, USA
| | | | - Jason Hinman
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Adam M Staffaroni
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Emily W Paolillo
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| |
Collapse
|
6
|
Edwards NC, Lao PJ, Alshikho MJ, Ericsson OM, Rizvi B, Petersen ME, O’Bryant S, Aguilar LF, Simoes S, Mapstone M, Tudorascu DL, Janelidze S, Hansson O, Handen BL, Christian BT, Lee JH, Lai F, Rosas HD, Zaman S, Lott IT, Yassa MA, Gutierrez J, Wilcock DM, Head E, Brickman AM. Cerebrovascular disease is associated with Alzheimer's plasma biomarker concentrations in adults with Down syndrome. Brain Commun 2024; 6:fcae331. [PMID: 39403075 PMCID: PMC11472828 DOI: 10.1093/braincomms/fcae331] [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: 05/03/2024] [Revised: 07/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
By age 40 years, over 90% of adults with Down syndrome have Alzheimer's disease pathology and most progress to dementia. Despite having few systemic vascular risk factors, individuals with Down syndrome have elevated cerebrovascular disease markers that track with the clinical progression of Alzheimer's disease, suggesting a role of cerebrovascular disease that is hypothesized to be mediated by inflammatory factors. This study examined the pathways through which small vessel cerebrovascular disease contributes to Alzheimer's disease-related pathophysiology and neurodegeneration in adults with Down syndrome. One hundred eighty-five participants from the Alzheimer's Biomarkers Consortium-Down Syndrome [mean (SD) age = 45.2 (9.3) years] with available MRI and plasma biomarker data were included in this study. White matter hyperintensity (WMH) volumes were derived from T2-weighted fluid-attenuated inversion recovery MRI scans, and plasma biomarker concentrations of amyloid beta 42/40, phosphorylated tau 217, astrocytosis (glial fibrillary acidic protein) and neurodegeneration (neurofilament light chain) were measured with ultrasensitive immunoassays. We examined the bivariate relationships of WMH, amyloid beta 42/40, phosphorylated tau 217 and glial fibrillary acidic protein with age-residualized neurofilament light chain across Alzheimer's disease diagnostic groups. A series of mediation and path analyses examined statistical pathways linking WMH and Alzheimer's disease pathophysiology to promote neurodegeneration in the total sample and groups stratified by clinical diagnosis. There was a direct and indirect bidirectional effect through the glial fibrillary acidic protein of WMH on phosphorylated tau 217 concentration, which was associated with neurofilament light chain concentration in the entire sample. Amongst cognitively stable participants, WMH was directly and indirectly, through glial fibrillary acidic protein, associated with phosphorylated tau 217 concentration, and in those with mild cognitive impairment, there was a direct effect of WMH on phosphorylated tau 217 and neurofilament light chain concentrations. There were no associations of WMH with biomarker concentrations among those diagnosed with dementia. The findings from this cross-sectional study suggest that among individuals with Down syndrome, cerebrovascular disease promotes neurodegeneration by increasing astrocytosis and tau pathophysiology in the presymptomatic phases of Alzheimer's disease, but future studies will need to confirm these associations with longitudinal data. This work joins an emerging literature that implicates cerebrovascular disease and its interface with neuroinflammation as a core pathological feature of Alzheimer's disease in adults with Down syndrome.
Collapse
Affiliation(s)
- Natalie C Edwards
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
- Department of Neuroscience, Columbia University, New York City, NY 10032, USA
| | - Patrick J Lao
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Mohamad J Alshikho
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Olivia M Ericsson
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Batool Rizvi
- Department of Neurobiology & Behavior, University of California, Irvine, CA 92697, USA
| | - Melissa E Petersen
- University of North Texas Health Science Center, Department of Pharmacology and Neuroscience, Fort Worth, TX 76107, USA
| | - Sid O’Bryant
- University of North Texas Health Science Center, Department of Pharmacology and Neuroscience, Fort Worth, TX 76107, USA
| | - Lisi Flores Aguilar
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA 92617, USA
| | - Sabrina Simoes
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund 221 00, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund 221 00, Sweden
- Memory Clinic, Skåne University Hospital, Malmö 214 28, Sweden
| | - Benjamin L Handen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Joseph H Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA
| | - H Diana Rosas
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Radiology, Center for Neuroimaging of Aging and Neurodegenerative Diseases, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
| | - Shahid Zaman
- Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ira T Lott
- Department of Pediatrics and Neurology, School of Medicine, University of California, Irvine, CA 92868, USA
| | - Michael A Yassa
- Department of Neurobiology & Behavior, University of California, Irvine, CA 92697, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697, USA
| | - José Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| | - Donna M Wilcock
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA 92617, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY 10032, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY 10032, USA
| |
Collapse
|
7
|
Morcillo‐Nieto AO, Zsadanyi SE, Arriola‐Infante JE, Carmona‐Iragui M, Montal V, Pegueroles J, Aranha MR, Vaqué‐Alcázar L, Padilla C, Benejam B, Videla L, Barroeta I, Fernandez S, Altuna M, Giménez S, González‐Ortiz S, Bargalló N, Ribas L, Arranz J, Torres S, Iulita MF, Belbin O, Camacho V, Alcolea D, Lleó A, Fortea J, Bejanin A. Characterization of white matter hyperintensities in Down syndrome. Alzheimers Dement 2024; 20:6527-6541. [PMID: 39087352 PMCID: PMC11497714 DOI: 10.1002/alz.14146] [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: 03/06/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION In Down syndrome (DS), white matter hyperintensities (WMHs) are highly prevalent, yet their topography and association with sociodemographic data and Alzheimer's disease (AD) biomarkers remain largely unexplored. METHODS In 261 DS adults and 131 euploid controls, fluid-attenuated inversion recovery magnetic resonance imaging scans were segmented and WMHs were extracted in concentric white matter layers and lobar regions. We tested associations with AD clinical stages, sociodemographic data, cerebrospinal fluid (CSF) AD biomarkers, and gray matter (GM) volume. RESULTS In DS, total WMHs arose at age 43 and showed stronger associations with age than in controls. WMH volume increased along the AD continuum, particularly in periventricular regions, and frontal, parietal, and occipital lobes. Associations were found with CSF biomarkers and temporo-parietal GM volumes. DISCUSSION WMHs increase 10 years before AD symptom onset in DS and are closely linked with AD biomarkers and neurodegeneration. This suggests a direct connection to AD pathophysiology, independent of vascular risks. HIGHLIGHTS White matter hyperintensities (WMHs) increased 10 years before Alzheimer's disease symptom onset in Down syndrome (DS). WMHs were strongly associated in DS with the neurofilament light chain biomarker. WMHs were more associated in DS with gray matter volume in parieto-temporal areas.
Collapse
|
8
|
Peavy GM, Võ N, Revta C, Lu AT, Lupo JL, Nam P, Nguyễn KH, Wang LS, Feldman HH. Asian Cohort for Alzheimer Disease (ACAD) Pilot Study: Vietnamese Americans. Alzheimer Dis Assoc Disord 2024; 38:277-284. [PMID: 39177172 PMCID: PMC11340683 DOI: 10.1097/wad.0000000000000631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/08/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The objective of this pilot study was to establish the feasibility of recruiting older Vietnamese Americans for research addressing genetic and nongenetic risk factors for Alzheimer disease (AD). METHODS Twenty-six Vietnamese Americans were recruited from communities in San Diego. A Community Advisory Board provided cultural and linguistic advice. Bilingual/bicultural staff measured neuropsychological, neuropsychiatric, lifestyle, and medical/neurological functioning remotely. Saliva samples allowed DNA extraction. A consensus team reviewed clinical data to determine a diagnosis of normal control (NC), mild cognitive impairment (MCI), or dementia. Exploratory analyses addressed AD risk by measuring subjective cognitive complaints (SCC), depression, and vascular risk factors (VRFs). RESULTS Twenty-five participants completed the study (mean age=73.8 y). Eighty percent chose to communicate in Vietnamese. Referrals came primarily from word of mouth within Vietnamese communities. Diagnoses included 18 NC, 3 MCI, and 4 dementia. Participants reporting SCC acknowledged more depressive symptoms and had greater objective cognitive difficulty than those without SCC. Eighty-eight percent of participants reported at least 1 VRF. DISCUSSION This pilot study supports the feasibility of conducting community-based research in older Vietnamese Americans. Challenges included developing linguistically and culturally appropriate cognitive and neuropsychiatric assessment tools. Exploratory analyses addressing nongenetic AD risk factors suggest topics for future study.
Collapse
Affiliation(s)
- Guerry M. Peavy
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
- Department of Neurosciences, Alzheimer’s Disease Research Center
| | - Namkhuê Võ
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
| | - Carolyn Revta
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
| | - Anna T. Lu
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
| | - Jody-Lynn Lupo
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
| | - Percival Nam
- Medical Student Y3, UC San Diego School of Medicine
| | - Khải H. Nguyễn
- Division of Geriatrics, Gerontology, and Palliative Care, UC San Diego School of Medicine, San Diego, CA
| | - Li-San Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Howard H. Feldman
- Department of Neurosciences, Alzheimer’s Disease Cooperative Study
- Department of Neurosciences, Alzheimer’s Disease Research Center
| |
Collapse
|
9
|
Yang HS, Yau WYW, Carlyle BC, Trombetta BA, Zhang C, Shirzadi Z, Schultz AP, Pruzin JJ, Fitzpatrick CD, Kirn DR, Rabin JS, Buckley RF, Hohman TJ, Rentz DM, Tanzi RE, Johnson KA, Sperling RA, Arnold SE, Chhatwal JP. Plasma VEGFA and PGF impact longitudinal tau and cognition in preclinical Alzheimer's disease. Brain 2024; 147:2158-2168. [PMID: 38315899 PMCID: PMC11146430 DOI: 10.1093/brain/awae034] [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: 08/16/2023] [Revised: 11/08/2023] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
Vascular dysfunction is increasingly recognized as an important contributor to the pathogenesis of Alzheimer's disease. Alterations in vascular endothelial growth factor (VEGF) pathways have been implicated as potential mechanisms. However, the specific impact of VEGF proteins in preclinical Alzheimer's disease and their relationships with other Alzheimer's disease and vascular pathologies during this critical early period remain to be elucidated. We included 317 older adults from the Harvard Aging Brain Study, a cohort of individuals who were cognitively unimpaired at baseline and followed longitudinally for up to 12 years. Baseline VEGF family protein levels (VEGFA, VEGFC, VEGFD, PGF and FLT1) were measured in fasting plasma using high-sensitivity immunoassays. Using linear mixed effects models, we examined the interactive effects of baseline plasma VEGF proteins and amyloid PET burden (Pittsburgh Compound-B) on longitudinal cognition (Preclinical Alzheimer Cognitive Composite-5). We further investigated if effects on cognition were mediated by early neocortical tau accumulation (flortaucipir PET burden in the inferior temporal cortex) or hippocampal atrophy. Lastly, we examined the impact of adjusting for baseline cardiovascular risk score or white matter hyperintensity volume. Baseline plasma VEGFA and PGF each showed a significant interaction with amyloid burden on prospective cognitive decline. Specifically, low VEGFA and high PGF were associated with greater cognitive decline in individuals with elevated amyloid, i.e. those on the Alzheimer's disease continuum. Concordantly, low VEGFA and high PGF were associated with accelerated longitudinal tau accumulation in those with elevated amyloid. Moderated mediation analyses confirmed that accelerated tau accumulation fully mediated the effects of low VEGFA and partially mediated (31%) the effects of high PGF on faster amyloid-related cognitive decline. The effects of VEGFA and PGF on tau and cognition remained significant after adjusting for cardiovascular risk score or white matter hyperintensity volume. There were concordant but non-significant associations with longitudinal hippocampal atrophy. Together, our findings implicate low VEGFA and high PGF in accelerating early neocortical tau pathology and cognitive decline in preclinical Alzheimer's disease. Additionally, our results underscore the potential of these minimally-invasive plasma biomarkers to inform the risk of Alzheimer's disease progression in the preclinical population. Importantly, VEGFA and PGF appear to capture distinct effects from vascular risks and cerebrovascular injury. This highlights their potential as new therapeutic targets, in combination with anti-amyloid and traditional vascular risk reduction therapies, to slow the trajectory of preclinical Alzheimer's disease and delay or prevent the onset of cognitive decline.
Collapse
Affiliation(s)
- Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Becky C Carlyle
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3PT, UK
| | - Bianca A Trombetta
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Can Zhang
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jeremy J Pruzin
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Banner Alzheimer’s Institute, Phoenix, AZ 85006, USA
| | | | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jennifer S Rabin
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Medicine, Rehabilitation Sciences Institute, University of Toronto, Toronto, ON M5G 1V7, Canada
| | - Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer’s Center, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Rudolph E Tanzi
- Harvard Medical School, Boston, MA 02115, USA
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Steven E Arnold
- Harvard Medical School, Boston, MA 02115, USA
- Alzheimer’s Clinical and Translational Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
10
|
Lao P, Edwards N, Flores-Aguilar L, Alshikho M, Rizvi B, Tudorascu D, Rosas HD, Yassa M, Christian BT, Mapstone M, Handen B, Zimmerman ME, Gutierrez J, Wilcock D, Head E, Brickman AM. Cerebrovascular disease emerges with age and Alzheimer's disease in adults with Down syndrome. Sci Rep 2024; 14:12334. [PMID: 38811657 PMCID: PMC11137035 DOI: 10.1038/s41598-024-61962-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
Adults with Down syndrome have a genetic form of Alzheimer's disease (AD) and evidence of cerebrovascular disease across the AD continuum, despite few systemic vascular risk factors. The onset and progression of AD in Down syndrome is highly age-dependent, but it is unknown at what age cerebrovascular disease emerges and what factors influence its severity. In the Alzheimer's Biomarker Consortium-Down Syndrome study (ABC-DS; n = 242; age = 25-72), we estimated the age inflection point at which MRI-based white matter hyperintensities (WMH), enlarged perivascular spaces (PVS), microbleeds, and infarcts emerge in relation to demographic data, risk factors, amyloid and tau, and AD diagnosis. Enlarged PVS and infarcts appear to develop in the early 30s, while microbleeds, WMH, amyloid, and tau emerge in the mid to late 30s. Age-residualized WMH were higher in women, in individuals with dementia, and with lower body mass index. Participants with hypertension and APOE-ε4 had higher age-residualized PVS and microbleeds, respectively. Lifespan trajectories demonstrate a dramatic cerebrovascular profile in adults with Down syndrome that appears to evolve developmentally in parallel with AD pathophysiology approximately two decades prior to dementia symptoms.
Collapse
Affiliation(s)
- Patrick Lao
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, PS Box 16, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Natalie Edwards
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, PS Box 16, New York, NY, 10032, USA
| | - Lisi Flores-Aguilar
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, 92697, USA
| | - Mohamad Alshikho
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, PS Box 16, New York, NY, 10032, USA
| | - Batool Rizvi
- Department of Neurology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Dana Tudorascu
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - H Diana Rosas
- Department of Neurology, Massachusetts General Hospital, Harvard Medical Center, Boston, MA, 02114, USA
| | - Michael Yassa
- Department of Neurology, University of California, Irvine, Irvine, CA, 92697, USA
| | | | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Benjamin Handen
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | | | - Jose Gutierrez
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Donna Wilcock
- Departments of Neurology and Anatomy, Cell Biology, and Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
| | - Adam M Brickman
- Gertrude H. Sergievsky Center and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, PS Box 16, New York, NY, 10032, USA.
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
11
|
Rizvi B, Lao PJ, Sathishkumar M, Taylor L, Queder N, McMillan L, Edwards NC, Keator DB, Doran E, Hom C, Nguyen D, Rosas HD, Lai F, Schupf N, Gutierrez J, Silverman W, Lott IT, Mapstone M, Wilcock DM, Head E, Yassa MA, Brickman AM. A pathway linking pulse pressure to dementia in adults with Down syndrome. Brain Commun 2024; 6:fcae157. [PMID: 38764776 PMCID: PMC11099660 DOI: 10.1093/braincomms/fcae157] [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: 11/07/2023] [Revised: 04/03/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Adults with Down syndrome are less likely to have hypertension than neurotypical adults. However, whether blood pressure measures are associated with brain health and clinical outcomes in this population has not been studied in detail. Here, we assessed whether pulse pressure is associated with markers of cerebrovascular disease and is linked to a diagnosis of dementia in adults with Down syndrome via structural imaging markers of cerebrovascular disease and atrophy. The study included participants with Down syndrome from the Alzheimer's Disease - Down Syndrome study (n = 195, age = 50.6 ± 7.2 years, 44% women, 18% diagnosed with dementia). Higher pulse pressure was associated with greater global, parietal and occipital white matter hyperintensity volume but not with enlarged perivascular spaces, microbleeds or infarcts. Using a structural equation model, we found that pulse pressure was associated with greater white matter hyperintensity volume, which in turn was related to increased neurodegeneration, and subsequent dementia diagnosis. Pulse pressure is an important determinant of brain health and clinical outcomes in individuals with Down syndrome despite the low likelihood of frank hypertension.
Collapse
Affiliation(s)
- Batool Rizvi
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Patrick J Lao
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Mithra Sathishkumar
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Lisa Taylor
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Nazek Queder
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Liv McMillan
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Natalie C Edwards
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - David B Keator
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, Orange, CA 92688, USA
| | - Christy Hom
- Department of Pediatrics, University of California, Irvine, Orange, CA 92688, USA
| | - Dana Nguyen
- Department of Pediatrics, University of California, Irvine, Orange, CA 92688, USA
| | - H Diana Rosas
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA 02114, USA
- Department of Radiology, Athinoula Martinos Center, Massachusetts General Hospital, Harvard University, Charlestown, MA 02129, USA
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA 02114, USA
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Jose Gutierrez
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Wayne Silverman
- Department of Pediatrics, University of California, Irvine, Orange, CA 92688, USA
| | - Ira T Lott
- Department of Pediatrics, University of California, Irvine, Orange, CA 92688, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, Irvine, CA 92697, USA
| | - Donna M Wilcock
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael A Yassa
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
12
|
Cha WJ, Yi D, Ahn H, Byun MS, Chang YY, Choi JM, Kim K, Choi H, Jung G, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Association between brain amyloid deposition and longitudinal changes of white matter hyperintensities. Alzheimers Res Ther 2024; 16:50. [PMID: 38454444 PMCID: PMC10918927 DOI: 10.1186/s13195-024-01417-8] [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: 11/04/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence suggests that not only cerebrovascular disease but also Alzheimer's disease (AD) pathological process itself cause cerebral white matter degeneration, resulting in white matter hyperintensities (WMHs). Some preclinical evidence also indicates that white matter degeneration may precede or affect the development of AD pathology. This study aimed to clarify the direction of influence between in vivo AD pathologies, particularly beta-amyloid (Aβ) and tau deposition, and WMHs through longitudinal approach. METHODS Total 282 older adults including cognitively normal and cognitively impaired individuals were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. The participants underwent comprehensive clinical and neuropsychological assessment, [11C] Pittsburgh Compound B PET for measuring Aβ deposition, [18F] AV-1451 PET for measuring tau deposition, and MRI scans with fluid-attenuated inversion recovery image for measuring WMH volume. The relationships between Aβ or tau deposition and WMH volume were examined using multiple linear regression analysis. In this analysis, baseline Aβ or tau were used as independent variables, and change of WMH volume over 2 years was used as dependent variable to examine the effect of AD pathology on increase of WMH volume. Additionally, we set baseline WMH volume as independent variable and longitudinal change of Aβ or tau deposition for 2 years as dependent variables to investigate whether WMH volume could precede AD pathologies. RESULTS Baseline Aβ deposition, but not tau deposition, had significant positive association with longitudinal change of WMH volume over 2 years. Baseline WMH volume was not related with any of longitudinal change of Aβ or tau deposition for 2 years. We also found a significant interaction effect between baseline Aβ deposition and sex on longitudinal change of WMH volume. Subsequent subgroup analyses showed that high baseline Aβ deposition was associated with increase of WMH volume over 2 years in female, but not in male. CONCLUSIONS Our findings suggest that Aβ deposition accelerates cerebral WMHs, particularly in female, whereas white matter degeneration appears not influence on longitudinal Aβ increase. The results also did not support any direction of influence between tau deposition and WMHs.
Collapse
Affiliation(s)
- Woo-Jin Cha
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jung-Min Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Royse SK, Snitz BE, Hengenius JB, Huppert TJ, Roush RE, Ehrenkranz RE, Wilson JD, Bertolet M, Reese AC, Cisneros G, Potopenko K, Becker JT, Cohen AD, Shaaban CE. Unhealthy white matter connectivity, cognition, and racialization in older adults. Alzheimers Dement 2024; 20:1483-1496. [PMID: 37828730 PMCID: PMC10947965 DOI: 10.1002/alz.13494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION White matter hyperintensities (WMH) may promote clinical Alzheimer's disease (AD) disparities between Black American (BA) and non-Hispanic White (nHW) populations. Using a novel measurement, unhealthy white matter connectivity (UWMC), we interrogated racialized group differences in associations between WMH in AD pathology-affected regions and cognition. METHODS UWMC is the proportion of white matter fibers that pass through WMH for every pair of brain regions. Individual regression models tested associations of UWMC in beta-amyloid (Aβ) or tau pathology-affected regions with cognition overall, stratified by racialized group, and with a racialized group interaction. RESULTS In 201 older adults ranging from cognitively unimpaired to AD, BA participants exhibited greater UWMC and worse cognition than nHW participants. UWMC was negatively associated with cognition in 17 and 5 Aβ- and tau-affected regions, respectively. Racialization did not modify these relationships. DISCUSSION Differential UWMC burden, not differential UWMC-and-cognition associations, may drive clinical AD disparities between racialized groups. HIGHLIGHTS Unhealthy white matter connectivity (UWMC) in Alzheimer's disease (AD) pathology-affected brain regions is associated with cognition. Relationships between UWMC and cognition are similar between Black American (BA) and non-Hispanic White (nHW) individuals. More UWMC may partially drive higher clinical AD burden in BA versus nHW populations. UWMC risk factors, particularly social and environmental, should be identified.
Collapse
Affiliation(s)
- Sarah K. Royse
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beth E. Snitz
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James B. Hengenius
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Theodore J. Huppert
- Department of Electrical EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rebecca E. Roush
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - James D. Wilson
- Department of Mathematics and StatisticsUniversity of San FranciscoSan FranciscoCaliforniaUSA
| | - Marnie Bertolet
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Geraldine Cisneros
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Katey Potopenko
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James T. Becker
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ann D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
14
|
Morales CD, Cotton-Samuel D, Lao PJ, Chang JF, Pyne JD, Alshikho MJ, Lippert RV, Bista K, Hale C, Edwards NC, Igwe KC, Deters K, Zimmerman ME, Brickman AM. Small vessel cerebrovascular disease is associated with cognition in prospective Alzheimer's clinical trial participants. Alzheimers Res Ther 2024; 16:25. [PMID: 38308344 PMCID: PMC10836014 DOI: 10.1186/s13195-024-01395-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: 09/11/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Secondary prevention clinical trials for Alzheimer's disease (AD) target amyloid accumulation in asymptomatic, amyloid-positive individuals, but it is unclear to what extent other pathophysiological processes, such as small vessel cerebrovascular disease, account for participant performance on the primary cognitive outcomes in those trials. White matter hyperintensities are areas of increased signal on T2-weighted magnetic resonance imaging (MRI) that reflect small vessel cerebrovascular disease. They are associated with cognitive functioning in older adults and with clinical presentation and course of AD, particularly when distributed in posterior brain regions. The purpose of this study was to examine to what degree regional WMH volume is associated with performance on the primary cognitive outcome measure in the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4) study, a secondary prevention trial. METHODS Data from 1791 participants (59.5% women, mean age (SD) 71.6 (4.74)) in the A4 study and the Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) companion study at the screening visit were used to quantify WMH volumes on T2-weighted fluid-attenuated inversion recovery (FLAIR) MR images. Cognition was assessed with the preclinical Alzheimer cognitive composite (PACC). We tested the association of total and regional WMH volumes with PACC performance, adjusting for age, education, and amyloid positivity status, with general linear models. We also considered interactions between WMH and amyloid positivity status. RESULTS Increased frontal and parietal lobe WMH volume was associated with poorer performance on the PACC. While amyloid positivity was also associated with lower cognitive test scores, WMH volumes did not interact with amyloid positivity status. CONCLUSION These results highlight the potential of small vessel cerebrovascular disease to drive AD-related cognitive profiles. Measures of small vessel cerebrovascular disease should be considered when evaluating outcome in trials, both as potential effect modifiers and as a possible target for intervention or prevention.
Collapse
Affiliation(s)
- Clarissa D Morales
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Dejania Cotton-Samuel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Patrick J Lao
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Julia F Chang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jeffrey D Pyne
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Mohamad J Alshikho
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Rafael V Lippert
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kelsang Bista
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Christiane Hale
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Natalie C Edwards
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kay C Igwe
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kacie Deters
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, USA
| | | | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Baena-Caldas GP, Li J, Pedraza L, Ghosh S, Kalmes A, Barone FC, Moreno H, Hernández AI. Neuroprotective effect of the RNS60 in a mouse model of transient focal cerebral ischemia. PLoS One 2024; 19:e0295504. [PMID: 38166102 PMCID: PMC10760892 DOI: 10.1371/journal.pone.0295504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 11/22/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Stroke is a major cause of death, disability, and public health problems. Its intervention is limited to early treatment with thrombolytics and/or endovascular clot removal with mechanical thrombectomy without any available subacute or chronic neuroprotective treatments. RNS60 has reduced neuroinflammation and increased neuronal survival in several animal models of neurodegeneration and trauma. The aim here was to evaluate whether RNS60 protects the brain and cognitive function in a mouse stroke model. METHODS Male C57BL/6J mice were subjected to sham or ischemic stroke surgery using 60-minute transient middle cerebral artery occlusion (tMCAo). In each group, mice received blinded daily administrations of RNS60 or control fluids (PNS60 or normal saline [NS]), beginning 2 hours after surgery over 13 days. Multiple neurobehavioral tests were conducted (Neurological Severity Score [mNSS], Novel Object Recognition [NOR], Active Place Avoidance [APA], and the Conflict Variant of APA [APAc]). On day 14, cortical microvascular perfusion (MVP) was measured, then brains were removed and infarct volume, immunofluorescence of amyloid beta (Aβ), neuronal density, microglial activation, and white matter damage/myelination were measured. SPSS was used for analysis (e.g., ANOVA for parametric data; Kruskal Wallis for non-parametric data; with post-hoc analysis). RESULTS Thirteen days of treatment with RNS60 reduced brain infarction, amyloid pathology, neuronal death, microglial activation, white matter damage, and increased MVP. RNS60 reduced brain pathology and resulted in behavioral improvements in stroke compared to sham surgery mice (increased memory-learning in NOR and APA, improved cognitive flexibility in APAc). CONCLUSION RNS60-treated mice exhibit significant protection of brain tissue and improved neurobehavioral functioning after tMCAo-stroke. Additional work is required to determine mechanisms, time-window of dosing, and multiple dosing volumes durations to support clinical stroke research.
Collapse
Affiliation(s)
- Gloria Patricia Baena-Caldas
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- Health Sciences Division, Department of Morphology, School of Biomedical Sciences, Universidad del Valle, Cali, Colombia
| | - Jie Li
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Lina Pedraza
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
| | - Supurna Ghosh
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Andreas Kalmes
- Revalesio Corporation, Tacoma, WA, United States of America
| | - Frank C. Barone
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - Herman Moreno
- Departments of Neurology and Physiology & Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - A. Iván Hernández
- Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, United States of America
- The Robert F. Furchgott Center for Neural and Behavioral Science, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| |
Collapse
|
16
|
Chum PP, Bishara MA, Solis SR, Behringer EJ. Cerebrovascular miRNAs Track Early Development of Alzheimer's Disease and Target Molecular Markers of Angiogenesis and Blood Flow Regulation. J Alzheimers Dis 2024; 99:S187-S234. [PMID: 37458037 PMCID: PMC10787821 DOI: 10.3233/jad-230300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Background Alzheimer's disease (AD) is associated with impaired cerebral circulation which underscores diminished delivery of blood oxygen and nutrients to and throughout the brain. In the 3xTg-AD mouse model, we have recently found that > 10 cerebrovascular miRNAs pertaining to vascular permeability, angiogenesis, and inflammation (e.g., let-7d, miR-99a, miR-132, miR-133a, miR-151-5p, and miR-181a) track early development of AD. Further, endothelial-specific miRNAs (miR-126-3p, miR-23a/b, miR-27a) alter with onset of overall AD pathology relative to stability of smooth muscle/pericyte-specific miRNAs (miR-143, miR-145). Objective We tested the hypothesis that cerebrovascular miRNAs indicating AD pathology share mRNA targets that regulate key endothelial cell functions such as angiogenesis, vascular permeability, and blood flow regulation. Methods As detected by NanoString nCounter miRNA Expression panel for 3xTg-AD mice, 61 cerebrovascular miRNAs and respective mRNA targets were examined using Ingenuity Pathway Analysis for canonical Cardiovascular (Cardio) and Nervous System (Neuro) Signaling. Results The number of targets regulated per miRNA were 21±2 and 33±3 for the Cardio and Neuro pathways respectively, whereby 14±2 targets overlap among pathways. Endothelial miRNAs primarily target members of the PDE, PDGF, SMAD, and VEGF families. Individual candidates regulated by≥4 miRNAs that best mark AD pathology presence in 3xTg-AD mice include CFL2, GRIN2B, PDGFB, SLC6A1, SMAD3, SYT3, and TNFRSF11B. Conclusion miRNAs selective for regulation of endothelial function and respective downstream mRNA targets support a molecular basis for dysregulated cerebral blood flow regulation coupled with enhanced cell growth, proliferation, and inflammation.
Collapse
Affiliation(s)
- Phoebe P. Chum
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | | | | | |
Collapse
|
17
|
Edwards NC, Lao PJ, Alshikho MJ, Ericsson OM, Rizvi B, Petersen ME, O’Bryant S, Flores-Aguilar L, Simoes S, Mapstone M, Tudorascu DL, Janelidze S, Hansson O, Handen BL, Christian BT, Lee JH, Lai F, Rosas HD, Zaman S, Lott IT, Yassa MA, Gutierrez J, Wilcock DM, Head E, Brickman AM. Cerebrovascular disease drives Alzheimer plasma biomarker concentrations in adults with Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.28.23298693. [PMID: 38076904 PMCID: PMC10705616 DOI: 10.1101/2023.11.28.23298693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Importance By age 40 years over 90% of adults with Down syndrome (DS) have Alzheimer's disease (AD) pathology and most progress to dementia. Despite having few systemic vascular risk factors, individuals with DS have elevated cerebrovascular disease (CVD) markers that track with the clinical progression of AD, suggesting a role for CVD that is hypothesized to be mediated by inflammatory factors. Objective To examine the pathways through which small vessel CVD contributes to AD-related pathophysiology and neurodegeneration in adults with DS. Design Cross sectional analysis of neuroimaging, plasma, and clinical data. Setting Participants were enrolled in Alzheimer's Biomarker Consortium - Down Syndrome (ABC-DS), a multisite study of AD in adults with DS. Participants One hundred eighty-five participants (mean [SD] age=45.2 [9.3] years) with available MRI and plasma biomarker data were included. White matter hyperintensity (WMH) volumes were derived from T2-weighted FLAIR MRI scans and plasma biomarker concentrations of amyloid beta (Aβ42/Aβ40), phosphorylated tau (p-tau217), astrocytosis (glial fibrillary acidic protein, GFAP), and neurodegeneration (neurofilament light chain, NfL) were measured with ultrasensitive immunoassays. Main Outcomes and Measures We examined the bivariate relationships of WMH, Aβ42/Aβ40, p-tau217, and GFAP with age-residualized NfL across AD diagnostic groups. A series of mediation and path analyses examined causal pathways linking WMH and AD pathophysiology to promote neurodegeneration in the total sample and groups stratified by clinical diagnosis. Results There was a direct and indirect bidirectional effect through GFAP of WMH on p-tau217 concentration, which was associated with NfL concentration in the entire sample. Among cognitively stable participants, WMH was directly and indirectly, through GFAP, associated with p-tau217 concentration, and in those with MCI, there was a direct effect of WMH on p-tau217 and NfL concentrations. There were no associations of WMH with biomarker concentrations among those diagnosed with dementia. Conclusions and Relevance The findings suggest that among individuals with DS, CVD promotes neurodegeneration by increasing astrocytosis and tau pathophysiology in the presymptomatic phases of AD. This work joins an emerging literature that implicates CVD and its interface with neuroinflammation as a core pathological feature of AD in adults with DS.
Collapse
Affiliation(s)
- Natalie C. Edwards
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
- Department of Neuroscience, Columbia University, New York City, NY, USA
| | - Patrick J. Lao
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Mohamad J. Alshikho
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Olivia M. Ericsson
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Batool Rizvi
- Department of Neurobiology & Behavior, University of California, Irvine, CA, USA
| | | | - Sid O’Bryant
- University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Lisi Flores-Aguilar
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA, USA
| | - Sabrina Simoes
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | | | - Joseph H. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Florence Lai
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - H Diana Rosas
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Center for Neuroimaging of Aging and neurodegenerative Diseases, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Shahid Zaman
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Ira T. Lott
- Department of Pediatrics and Neurology, School of Medicine, University of California, Irvine, CA, USA
| | - Michael A. Yassa
- Department of Neurobiology & Behavior, University of California, Irvine, CA, USA
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| | - José Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
- Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, University of California, Irvine, CA, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, NY, USA
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York City, NY, USA
| |
Collapse
|
18
|
Eloyan A, Thangarajah M, An N, Borowski BJ, Reddy AL, Aisen P, Dage JL, Foroud T, Ghetti B, Griffin P, Hammers D, Iaccarino L, Jack CR, Kirby K, Kramer J, Koeppe R, Kukull WA, La Joie R, Mundada NS, Murray ME, Nudelman K, Rumbaugh M, Soleimani-Meigooni DN, Toga A, Touroutoglou A, Atri A, Day GS, Duara R, Graff-Radford NR, Honig LS, Jones DT, Masdeu J, Mendez MF, Musiek E, Onyike CU, Rogalski E, Salloway S, Sha S, Turner RS, Wingo TS, Wolk DA, Womack K, Beckett L, Gao S, Carrillo MC, Rabinovici G, Apostolova LG, Dickerson B, Vemuri P. White matter hyperintensities are higher among early-onset Alzheimer's disease participants than their cognitively normal and early-onset nonAD peers: Longitudinal Early-onset Alzheimer's Disease Study (LEADS). Alzheimers Dement 2023; 19 Suppl 9:S89-S97. [PMID: 37491599 PMCID: PMC10808262 DOI: 10.1002/alz.13402] [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: 03/03/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION We compared white matter hyperintensities (WMHs) in early-onset Alzheimer's disease (EOAD) with cognitively normal (CN) and early-onset amyloid-negative cognitively impaired (EOnonAD) groups in the Longitudinal Early-Onset Alzheimer's Disease Study. METHODS We investigated the role of increased WMH in cognition and amyloid and tau burden. We compared WMH burden of 205 EOAD, 68 EOnonAD, and 89 CN participants in lobar regions using t-tests and analyses of covariance. Linear regression analyses were used to investigate the association between WMH and cognitive impairment and that between amyloid and tau burden. RESULTS EOAD showed greater WMHs compared with CN and EOnonAD participants across all regions with no significant differences between CN and EOnonAD groups. Greater WMHs were associated with worse cognition. Tau burden was positively associated with WMH burden in the EOAD group. DISCUSSION EOAD consistently showed higher WMH volumes. Overall, greater WMHs were associated with worse cognition and higher tau burden in EOAD. HIGHLIGHTS This study represents a comprehensive characterization of WMHs in sporadic EOAD. WMH volumes are associated with tau burden from positron emission tomography (PET) in EOAD, suggesting WMHs are correlated with increasing burden of AD. Greater WMH volumes are associated with worse performance on global cognitive tests. EOAD participants have higher WMH volumes compared with CN and early-onset amyloid-negative cognitively impaired (EOnonAD) groups across all brain regions.
Collapse
Affiliation(s)
- Ani Eloyan
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA, 02903
| | - Maryanne Thangarajah
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA, 02903
| | - Na An
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA, 02903
| | - Bret J. Borowski
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA, 55905
| | - Ashritha L. Reddy
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA, 55905
| | - Paul Aisen
- Alzheimer’s Therapeutic Research Institute, University of Southern California, San Diego, California, USA, 92121
| | - Jeffrey L. Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Bernardino Ghetti
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
- Department of Pathology & Laboratory Medicine Indiana University School of Medicine, Indianapolis, Indiana, USA, 02912
| | - Percy Griffin
- Medical & Scientific Relations Division, Alzheimer’s Association, Chicago, Illinois, USA, 60603
| | - Dustin Hammers
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Leonardo Iaccarino
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA, 94143
| | - Clifford R. Jack
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Kala Kirby
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Joel Kramer
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA, 94143
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA, 48109
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, Washington, USA, 98195
| | - Renaud La Joie
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA, 94143
| | - Nidhi S Mundada
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA, 94143
| | - Melissa E. Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA, 32224
| | - Kelly Nudelman
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Malia Rumbaugh
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | | | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, California, USA, 90033
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA, 02114
| | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, Arizona, USA, 85351
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA, 32224
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease and Memory Disorders, Mount Sinai Medical Center, Miami, Florida, USA, 33140
| | | | - Lawrence S. Honig
- Taub Institute and Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA,10032
| | - David T. Jones
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA, 55905
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA, 55905
| | - Joseph Masdeu
- Nantz National Alzheimer Center, Houston Methodist and Weill Cornell Medicine, Houston, Texas, USA, 77030
| | - Mario F. Mendez
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA, 90095
| | - Erik Musiek
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA, 63108
| | - Chiadi U. Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA, 21205
| | - Emily Rogalski
- Department of Psychiatry and Behavioral Sciences, Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA, 60611
| | - Stephen Salloway
- Department of Neurology, Alpert Medical School, Brown University, Providence, Rhode Island, USA, 02912
| | - Sharon Sha
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, California, USA, 94304
| | - Raymond S. Turner
- Department of Neurology, Georgetown University, Washington D.C., USA, 20007
| | - Thomas S. Wingo
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA, 30322
| | - David A. Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA, 19104
| | - Kyle Womack
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA, 63108
| | - Laurel Beckett
- Department of Public Health Sciences, University of California – Davis, Davis, California, USA, 95616
| | - Sujuan Gao
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
| | - Maria C. Carrillo
- Medical & Scientific Relations Division, Alzheimer’s Association, Chicago, Illinois, USA, 60603
| | - Gil Rabinovici
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA, 94143
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA, 46202
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine Indianapolis, Indianapolis, Indiana, USA, 46202
| | - Brad Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA, 02114
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA, 55905
| | | |
Collapse
|
19
|
Rizvi B, Lao PJ, Sathishkumar M, Taylor L, Queder N, McMillan L, Edwards N, Keator DB, Doran E, Hom C, Nguyen D, Rosas HD, Lai F, Schupf N, Gutierrez J, Silverman W, Lott IT, Mapstone M, Wilcock DM, Head E, Yassa MA, Brickman AM. Pathways linking pulse pressure to dementia in adults with Down syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.10.26.23297625. [PMID: 37961444 PMCID: PMC10635215 DOI: 10.1101/2023.10.26.23297625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Individuals with Down syndrome (DS) are less likely to have hypertension than neurotypical adults. However, whether blood pressure measures are associated with brain health and clinical outcomes in this population has not been studied in detail. Here, we assessed whether pulse pressure is associated with markers of cerebrovascular disease, entorhinal cortical atrophy, and diagnosis of dementia in adults with DS. Participants with DS from the Biomarkers of Alzheimer's Disease in Adults with Down Syndrome study (ADDS; n=195, age=50.6±7.2 years, 44% women, 18% diagnosed with dementia) were included. Higher pulse pressure was associated with greater global, parietal, and occipital WMH volume. Pulse pressure was not related to enlarged PVS, microbleeds, infarcts, entorhinal cortical thickness, or dementia diagnosis. However, in a serial mediation model, we found that pulse pressure was indirectly related to dementia diagnosis through parieto-occipital WMH and, subsequently through entorhinal cortical thickness. Higher pulse pressure may be a risk factor for dementia in people with DS by promoting cerebrovascular disease, which in turn affects neurodegeneration. Pulse pressure is an important determinant of brain health and clinical outcomes in individuals with Down syndrome despite the low likelihood of frank hypertension.
Collapse
Affiliation(s)
- Batool Rizvi
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Patrick J. Lao
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Mithra Sathishkumar
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Lisa Taylor
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Nazek Queder
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Liv McMillan
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Natalie Edwards
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - David B. Keator
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Eric Doran
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Christy Hom
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Dana Nguyen
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - H. Diana Rosas
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
- Department of Radiology, Athinoula Martinos Center, Massachusetts General Hospital, Harvard University, Charlestown, MA, USA
| | - Florence Lai
- Department of Neurology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jose Gutierrez
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Wayne Silverman
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Ira T. Lott
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA, USA
| | - Donna M. Wilcock
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Michael A. Yassa
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA
- Department of Neurobiology and Behavior, University of California, Irvine, CA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
20
|
Brickman AM, Rizvi B. White matter hyperintensities and Alzheimer's disease: An alternative view of an alternative hypothesis. Alzheimers Dement 2023; 19:4260-4261. [PMID: 37437028 PMCID: PMC11465501 DOI: 10.1002/alz.13371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 07/14/2023]
Abstract
White matter hyperintensities (WMH) - - areas of increased signal appearing on T2-weighted magnetic resonance imaging - - are associated with Alzheimer’s disease (AD) risk and progression in late onset and genetic forms. Although typically attributable to macrostructural damage due to small vessel cerebrovascular disease or dysfunction, in a paper by Garnier-Crussard and colleagues[1 ], the authors review recent work suggesting an additional Wallerian-like component to WMH and argue that the elevated WMH seen in AD is a downstream phenomenon secondary to neurodegeneration. Here, we maintain that consideration of pathological correlates, animal work, brain perfusion patterns, longitudinal and incident data, and vascular risk factors provides evidence that is not inconsistent with a vasculogenic source of WMH. Future studies inspired by the consistent observations linking WMH to AD are needed to continue to understand potential vascular contributions to the disease.
Collapse
Affiliation(s)
- Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain and Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168 Street, New York, NY 10032
| | - Batool Rizvi
- Center for the Neurobiology of Learning and Memory and Department of Neurobiology and Behavior, University of California, Irvine, 1400 Biological Sciences III, Irvine, CA, 92697
| |
Collapse
|
21
|
Garnier-Crussard A, Chételat G. White matter hyperintensities in Alzheimer's disease: Beyond (but not instead of) the vascular contribution. Alzheimers Dement 2023; 19:4262-4263. [PMID: 37437034 DOI: 10.1002/alz.13372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 07/14/2023]
Affiliation(s)
- Antoine Garnier-Crussard
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders,", Neuropresage Team, Cyceron, France
- Clinical and Research Memory Center of Lyon, Lyon Institute for Aging, Hospices Civils de Lyon, Villeurbanne, France
| | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders,", Neuropresage Team, Cyceron, France
| |
Collapse
|
22
|
Garnier-Crussard A, Cotton F, Krolak-Salmon P, Chételat G. White matter hyperintensities in Alzheimer's disease: Beyond vascular contribution. Alzheimers Dement 2023; 19:3738-3748. [PMID: 37027506 DOI: 10.1002/alz.13057] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/15/2023] [Accepted: 03/03/2023] [Indexed: 04/09/2023]
Abstract
White matter hyperintensities (WMH), frequently seen in older adults, are usually considered vascular lesions, and participate in the vascular contribution to cognitive impairment and dementia. However, emerging evidence highlights the heterogeneity of WMH pathophysiology, suggesting that non-vascular mechanisms could also be involved, notably in Alzheimer's disease (AD). This led to the alternative hypothesis that in AD, part of WMH may be secondary to AD-related processes. The current perspective brings together the arguments from different fields of research, including neuropathology, neuroimaging and fluid biomarkers, and genetics, in favor of this alternative hypothesis. Possible underlying mechanisms leading to AD-related WMH, such as AD-related neurodegeneration or neuroinflammation, are discussed, as well as implications for diagnostic criteria and management of AD. We finally discuss ways to test this hypothesis and remaining challenges. Acknowledging the heterogeneity of WMH and the existence of AD-related WMH may improve personalized diagnosis and care of patients.
Collapse
Affiliation(s)
- Antoine Garnier-Crussard
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Neuropresage Team, Cyceron, Caen, France
- Clinical and Research Memory Center of Lyon, Lyon Institute For Aging, Hospices Civils de Lyon, Villeurbanne, France
- Eduwell team, Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, UCBL1, Lyon, France
| | - François Cotton
- Radiology Department, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
- CREATIS, INSERM U1044, CNRS UMR 5220, UCBL1, Villeurbanne, France
| | - Pierre Krolak-Salmon
- Clinical and Research Memory Center of Lyon, Lyon Institute For Aging, Hospices Civils de Lyon, Villeurbanne, France
- Eduwell team, Lyon Neuroscience Research Center (CRNL), INSERM U1028, CNRS UMR5292, UCBL1, Lyon, France
| | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Neuropresage Team, Cyceron, Caen, France
| |
Collapse
|
23
|
Yao Q, Jiang K, Lin F, Zhu T, Khan NH, Jiang E. Pathophysiological Association of Alzheimer's Disease and Hypertension: A Clinical Concern for Elderly Population. Clin Interv Aging 2023; 18:713-728. [PMID: 37181536 PMCID: PMC10167960 DOI: 10.2147/cia.s400527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/22/2023] [Indexed: 05/16/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia and the fifth leading cause of death in the adult population has a complex pathophysiological link with hypertension (HTN). A growing volume of published literature on a parallel elevation of blood pressure (BP), amyloid plaques, and neurofibrillary tangles formation in post-middle of human brain cells has developed new, widely accepting foundations on this association. In particular, HTN in elderly life mediates cerebral blood flow dysfunction, neuronal dysfunction, and significant decline in cognitive impairment, primarily in the late-life populace, governing the onset of AD. Thus, HTN is an established risk factor for AD. Considering the impact of AD, 1.89 million deaths annually, and the failure of palliative therapies to cure AD, the scientific research community is looking to adopt integrated approaches to target early modified risk factors like HTN to reduce AD burden. The current review highlights the significance and impact of HTN-based prevention in lowering the AD burden in the elderly by providing a comprehensive overview of the physiological relationship between AD and HTN with an in-detail explanation of the role and applications of pathological biomarkers in this clinical association. The review will gain worth in presenting new insights and providing inclusive discussion on the correlation between HTN and cognitive impairment. It will increase across a wider scientific audience to expand understanding of this pathophysiological association.
Collapse
Affiliation(s)
- Qianqian Yao
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
| | - Kexin Jiang
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
| | - Fei Lin
- School of Medicine, Shangqiu Institute of Technology, Shangqiu, People’s Republic of China
| | - Tao Zhu
- Department of Geriatrics, Kaifeng Traditional Chinese Medicine Hospital, Kaifeng, People’s Republic of China
| | - Nazeer Hussain Khan
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, People’s Republic of China
| | - Enshe Jiang
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, People’s Republic of China
| |
Collapse
|
24
|
Zhao J, Wang X, Yu M, Zhang S, Li Q, Liu H, Zhang J, Cai R, Lu C, Li S. The Relevance of Serum Macrophage Migration Inhibitory Factor Level and Executive Function in Patients with White Matter Hyperintensity in Cerebral Small Vessel Disease. Brain Sci 2023; 13:brainsci13040616. [PMID: 37190581 DOI: 10.3390/brainsci13040616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
(1) Objective: To investigate the relationship between serum macrophage migration inhibitory factor (MIF) level and white matter hyperintensity (WMH) and executive function (EF) in cerebral small vascular disease (CSVD), and assess the impact and predictive value of MIF level and Fazekas scores in CSVD-related cognitive impairment (CI) (CSVD-CI); (2) Methods: A total of 117 patients with WMH admitted to the First Affiliated Hospital of Xinxiang Medical College from January 2022 to August 2022 were enrolled. According to the Montreal cognitive assessment (MoCA) scale, subjects were divided into a normal cognitive group and an impaired group. All subjects required serum MIF level, 3.0 T MRI, and neuropsychological evaluation to investigate the risk factors for CDVD-CI, analyze the correlation between MIF level, WMH, and EF, and to analyze the diagnostic value of MIF and WMH degree in predicting CSVD-CI; (3) Results: 1. Fazekas score and MIF level were the risk factors of CSVD-CI. 2. The Fazekas score was negatively correlated with MoCA score, positively correlated with Stroop C-Time, Stroop C-Mistake, Stroop interference effects (SIE)-Time, SIE-Mistake, and color trails test (CTT) interference effects (CIE) (B-A). 3. The MIF level was positively correlated with Fazekas score, Stroop C-Time, SIE-Time, CTT B-Time, and CIE (B-A), and negatively correlated with MoCA score. 4. Fazekas score and MIF level were significant factors for diagnosing CSVD-CI; (4) Conclusion: The Fazekas score and MIF level may be the risk factors of CSVD-CI, and they are closely correlated to CI, especially the EF, and they have diagnostic value for CSVD-CI.
Collapse
Affiliation(s)
- Jianhua Zhao
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Xiaoting Wang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Miao Yu
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Shiyun Zhang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Qiong Li
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Hao Liu
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Jian Zhang
- Imaging Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Ruiyan Cai
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Neurology Department, First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453100, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang 453003, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Kapadia A, Billimoria K, Desai P, Grist JT, Heyn C, Maralani P, Symons S, Zaccagna F. Hypoperfusion Precedes Tau Deposition in the Entorhinal Cortex: A Retrospective Evaluation of ADNI-2 Data. J Clin Neurol 2023; 19:131-137. [PMID: 36647226 PMCID: PMC9982189 DOI: 10.3988/jcn.2022.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Tau deposition in the entorhinal cortex is the earliest pathological feature of Alzheimer's disease (AD). However, this feature has also been observed in cognitively normal (CN) individuals and those with mild cognitive impairment (MCI). The precise pathophysiology for the development of tau deposition remains unclear. We hypothesized that reduced cerebral perfusion is associated with the development of tau deposition. METHODS A subset of the Alzheimer's Disease Neuroimaging Initiative data set was utilized. Included patients had undergone arterial spin labeling perfusion MRI along with [18F]flortaucipir tau PET at baseline, within 1 year of the MRI, and a follow-up at 6 years. The association between baseline cerebral blood flow (CBF) and the baseline and 6-year tau PET was assessed. Univariate and multivariate linear modeling was performed, with p<0.05 indicating significance. RESULTS Significant differences were found in the CBF between patients with AD and MCI, and CN individuals in the left entorhinal cortex (p=0.013), but not in the right entorhinal cortex (p=0.076). The difference in maximum standardized uptake value ratio between 6 years and baseline was significantly and inversely associated with the baseline mean CBF (p=0.042, R²=0.54) in the left entorhinal cortex but not the right entorhinal cortex. Linear modeling demonstrated that CBF predicted 6-year tau deposition (p=0.015, R²=0.11). CONCLUSIONS The results of this study suggest that a reduction in CBF at the entorhinal cortex precedes tau deposition. Further work is needed to understand the mechanism underlying tau deposition in aging and disease.
Collapse
Affiliation(s)
- Anish Kapadia
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| | - Krish Billimoria
- MD Program, Temetry Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Prarthna Desai
- Department of Medicine, Maharaja Sayajirao University of Baroda, Vadodara, India
| | - James T. Grist
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK.,Department of Radiology, Oxford University Hospitals Trust, Oxford, UK.,Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Chris Heyn
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Pejman Maralani
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sean Symons
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada.,Division of Neuroradiology, Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Fulvio Zaccagna
- Division of Neuroradiology, Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Zhao J, Wang X, Li Q, Lu C, Li S. The relevance of serum macrophage migratory inhibitory factor and cognitive dysfunction in patients with cerebral small vascular disease. Front Aging Neurosci 2023; 15:1083818. [PMID: 36824264 PMCID: PMC9941340 DOI: 10.3389/fnagi.2023.1083818] [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: 10/29/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Cerebral small vascular disease (CSVD) is a common type of cerebrovascular disease, and an important cause of vascular cognitive impairment (VCI) and stroke. The disease burden is expected to increase further as a result of population aging, an ongoing high prevalence of risk factors (e.g., hypertension), and inadequate management. Due to the poor understanding of pathophysiology in CSVD, there is no effective preventive or therapeutic approach for CSVD. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that is related to the occurrence and development of vascular dysfunction diseases. Therefore, MIF may contribute to the pathogenesis of CSVD and VCI. Here, reviewed MIF participation in chronic cerebral ischemia-hypoperfusion and neurodegeneration pathology, including new evidence for CSVD, and its potential role in protection against VCI.
Collapse
Affiliation(s)
- Jianhua Zhao
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China,*Correspondence: Jianhua Zhao,
| | - Xiaoting Wang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qiong Li
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
27
|
Mehta RI, Mehta RI. The Vascular-Immune Hypothesis of Alzheimer's Disease. Biomedicines 2023; 11:408. [PMID: 36830944 PMCID: PMC9953491 DOI: 10.3390/biomedicines11020408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and irreversible neurodegenerative disorder with unknown etiology. While its cause is unclear, a number of theories have been proposed to explain the pathogenesis of AD. In large part, these have centered around potential causes for intracerebral accumulation of beta-amyloid (βA) and tau aggregates. Yet, persons with AD dementia often exhibit autopsy evidence of mixed brain pathologies including a myriad of vascular changes, vascular brain injuries, complex brain inflammation, and mixed protein inclusions in addition to hallmark neuropathologic lesions of AD, namely insoluble βA plaques and neurofibrillary tangles (NFTs). Epidemiological data demonstrate that overlapping lesions diminish the βA plaque and NFT threshold necessary to precipitate clinical dementia. Moreover, a subset of persons who exhibit AD pathology remain resilient to disease while other persons with clinically-defined AD dementia do not exhibit AD-defining neuropathologic lesions. It is increasingly recognized that AD is a pathologically heterogeneous and biologically multifactorial disease with uncharacterized biologic phenomena involved in its genesis and progression. Here, we review the literature with regard to neuropathologic criteria and incipient AD changes, and discuss converging concepts regarding vascular and immune factors in AD.
Collapse
Affiliation(s)
- Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Rupal I. Mehta
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
28
|
Jiang X, O'Bryant SE, Johnson LA, Rissman RA, Yaffe K. Association of cardiovascular risk factors and blood biomarkers with cognition: The HABS-HD study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12394. [PMID: 36911361 PMCID: PMC9994160 DOI: 10.1002/dad2.12394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/08/2022] [Accepted: 12/20/2022] [Indexed: 03/14/2023]
Abstract
Introduction To determine if cardiovascular risk factor (CVRF) burden is associated with Alzheimer's disease (AD) biomarkers and whether they synergistically associate with cognition. Methods We cross-sectionally studied 1521 non-demented Mexican American (52%) and non-Hispanic White individuals aged ≥50 years. A composite score was calculated by averaging the z-scores of five cognitive tests. Plasma β-amyloid (Aβ) 42/40, total tau (t-tau), and neurofilament light (NfL) were assayed using Simoa. CVRF burden was assessed using the Framingham Risk Score (FRS). Results Compared to low FRS (< 10% risk), high FRS (≥ 20% risk) was independently associated with increased t-tau and NfL. High FRS was significantly associated with higher NfL only among Mexican American individuals. Intermediate or high FRS (vs. low FRS) were independently associated with lower cognition, and the association remained significant after adjusting for plasma biomarkers. Hypertension synergistically interacted with t-tau and NfL (p < 0.05). Discussion CVRFs play critical roles, both through independent and neurodegenerative pathways, on cognition.
Collapse
Affiliation(s)
- Xiaqing Jiang
- Department of Psychiatry and Behavioral SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sid E. O'Bryant
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Leigh A. Johnson
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
- Veterans Affairs San Diego Healthcare SystemSan DiegoCaliforniaUSA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
- San Francisco VA Health Care SystemSan FranciscoCaliforniaUSA
| | | |
Collapse
|
29
|
Hakim MA, Behringer EJ. K IR channel regulation of electrical conduction along cerebrovascular endothelium: Enhanced modulation during Alzheimer's disease. Microcirculation 2023; 30:e12797. [PMID: 36577656 PMCID: PMC9885900 DOI: 10.1111/micc.12797] [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: 07/15/2022] [Revised: 11/22/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Endothelial cell (EC) coupling occurs through gap junctions and underlies cerebral blood flow regulation governed by inward-rectifying K+ (KIR ) channels. This study addressed effects of KIR channel activity on EC coupling before and during Alzheimer's disease (AD). METHODS Intact EC tubes (width: ~90-100 μm; length: ~0.5 mm) were freshly isolated from posterior cerebral arteries of young Pre-AD (1-3 months) and aged AD (13-18 months) male and female 3xTg-AD mice. Dual intracellular microelectrodes applied simultaneous current injections (±0.5-3 nA) and membrane potential (Vm ) recordings in ECs at distance ~400 μm. Elevated extracellular potassium ([K+ ]E ; 8-15 mmol/L; reference, 5 mmol/L) activated KIR channels. RESULTS Conducted Vm (∆Vm ) responses ranged from ~-30 to 30 mV in response to -3 to +3 nA (linear regression, R2 ≥ .99) while lacking rectification for charge polarity or axial direction of spread. Conduction slope decreased ~10%-20% during 15 mmol/L [K+ ]E in Pre-AD males and AD females. 15 mmol/L [K+ ]E decreased conduction by ~10%-20% at lower ∆Vm thresholds in AD animals (~±20 mV) versus Pre-AD (~±25 mV). AD increased conducted hyperpolarization by ~10%-15% during 8-12 mmol/L [K+ ]E . CONCLUSIONS Brain endothelial KIR channel activity modulates bidirectional spread of vasoreactive signals with enhanced regulation of EC coupling during AD pathology.
Collapse
Affiliation(s)
- Md A. Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Erik J. Behringer
- Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA,Corresponding Author: Erik J. Behringer, Ph.D., Department of Basic Sciences, 11041 Campus Street, Risley Hall, Loma Linda University, Loma Linda, CA 92350, , tel: (909) 651-5334, fax: (909) 558-0119
| |
Collapse
|
30
|
Hart G, Huang CW, Rust N, Wu HF. Altered O-GlcNAcylation and mitochondrial dysfunction, a molecular link between brain glucose dysregulation and sporadic Alzheimer’s disease. Neural Regen Res 2023; 18:779-783. [DOI: 10.4103/1673-5374.354515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Newton P, Tchounguen J, Pettigrew C, Lim C, Lin Z, Lu H, Moghekar A, Albert M, Soldan A. Regional White Matter Hyperintensities and Alzheimer's Disease Biomarkers Among Older Adults with Normal Cognition and Mild Cognitive Impairment. J Alzheimers Dis 2023; 92:323-339. [PMID: 36744337 PMCID: PMC10041440 DOI: 10.3233/jad-220846] [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: 12/29/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) frequently co-occurs with other brain pathologies. Recent studies suggest there may be a mechanistic link between AD and small vessel cerebrovascular disease (CVD), as opposed to simply the overlap of two disorders. OBJECTIVE We investigated the cross-sectional relationship between white matter hyperintensity (WMH) volumes (markers of CVD) and cerebrospinal fluid (CSF) biomarkers of AD. METHODS WMH volumes were assessed globally and regionally (i.e., frontal, parietal, temporal, occipital, and limbic). CSF AD biomarkers (i.e., Aβ 40, Aβ 42, Aβ 42/Aβ 40 ratio, phosphorylated tau-181 [p-tau181], and total tau [t-tau]) were measured among 152 non-demented individuals (134 cognitively unimpaired and 18 with mild cognitive impairment (MCI)). RESULTS Linear regression models showed that among all subjects, higher temporal WHM volumes were associated with AD biomarkers (higher levels of p-tau181, t-tau, and Aβ 40), particularly among APOE ɛ 4 carriers (independent of Aβ 42 levels). Higher vascular risk scores were associated with greater parietal and frontal WMH volumes (independent of CSF AD biomarker levels). Among subjects with MCI only, parietal WMH volumes were associated with a lower level of Aβ 42/Aβ 40. In addition, there was an association between higher global WMH volumes and higher CSF t-tau levels among younger participants versus older ones (∼<65 versus 65+ years), independent of Aβ 42/Aβ 40 and p-tau181. CONCLUSION These findings suggest that although WMH are primarily related to systemic vascular risk and neurodegeneration (i.e., t-tau), AD-specific pathways may contribute to the formation of WMH in a regionally-specific manner, with neurofibrillary tangles (i.e., p-tau) playing a role in temporal WMHs and amyloid (i.e., Aβ 42/Aβ 40) in parietal WMHs.
Collapse
Affiliation(s)
- Princess Newton
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | | | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chantelle Lim
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zixuan Lin
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - the BIOCARD Research Team
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Schoemaker D, Zanon Zotin MC, Chen K, Igwe KC, Vila-Castelar C, Martinez J, Baena A, Fox-Fuller JT, Lopera F, Reiman EM, Brickman AM, Quiroz YT. White matter hyperintensities are a prominent feature of autosomal dominant Alzheimer’s disease that emerge prior to dementia. Alzheimers Res Ther 2022; 14:89. [PMID: 35768838 PMCID: PMC9245224 DOI: 10.1186/s13195-022-01030-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/06/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
To promote the development of effective therapies, there is an important need to characterize the full spectrum of neuropathological changes associated with Alzheimer’s disease. In line with this need, this study examined white matter abnormalities in individuals with early-onset autosomal dominant Alzheimer’s disease, in relation to age and symptom severity.
Methods
This is a cross-sectional analysis of data collected in members of a large kindred with a PSEN1 E280A mutation. Participants were recruited between September 2011 and July 2012 from the Colombian Alzheimer’s Prevention Initiative registry. The studied cohort comprised 50 participants aged between 20 and 55 years, including 20 cognitively unimpaired mutation carriers, 9 cognitively impaired mutation carriers, and 21 non-carriers. Participants completed an MRI, a lumbar puncture for cerebrospinal fluid collection, a florbetapir PET scan, and neurological and neuropsychological examinations. The volume of white matter hyperintensities (WMH) was compared between cognitively unimpaired carriers, cognitively impaired carriers, and non-carriers. Relationships between WMH, age, and cognitive performance were further examined in mutation carriers.
Results
The mean (SD) age of participants was 35.8 (9.6) years and 64% were women. Cardiovascular risk factors were uncommon and did not differ across groups. Cognitively impaired carriers [median, 6.37; interquartile range (IQR), 9.15] had an increased volume of WMH compared to both cognitively unimpaired carriers [median, 0.85; IQR, 0.79] and non-carriers [median, 1.07; IQR, 0.71]. In mutation carriers, the volume of WMH strongly correlated with cognition and age, with evidence for an accelerated rate of changes after the age of 43 years, 1 year earlier than the estimated median age of symptom onset. In multivariable regression models including cortical amyloid retention, superior parietal lobe cortical thickness, and cerebrospinal fluid phospho-tau, the volume of WMH was the only biomarker independently and significantly contributing to the total explained variance in cognitive performance.
Conclusions
The volume of WMH is increased among individuals with symptomatic autosomal-dominant Alzheimer’s disease, begins to increase prior to clinical symptom onset, and is an independent determinant of cognitive performance in this group. These findings suggest that WMH are a key component of autosomal-dominant Alzheimer’s disease that is closely related to the progression of clinical symptoms.
Collapse
|
33
|
Abdulrahman H, van Dalen JW, den Brok M, Latimer CS, Larson EB, Richard E. Hypertension and Alzheimer's disease pathology at autopsy: A systematic review. Alzheimers Dement 2022; 18:2308-2326. [PMID: 35758526 PMCID: PMC9796086 DOI: 10.1002/alz.12707] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/27/2022] [Accepted: 04/27/2022] [Indexed: 01/31/2023]
Abstract
Hypertension is an important risk factor for Alzheimer's disease (AD) and all-cause dementia. The mechanisms underlying this association are unclear. Hypertension may be associated with AD neuropathological changes (ADNC), but reports are sparse and inconsistent. This systematic review included 15 autopsy studies (n = 5879) from observational cohorts. Studies were highly heterogeneous regarding populations, follow-up duration, hypertension operationalization, neuropathological methods, and statistical analyses. Hypertension seems associated with higher plaque and tangle burden, but results are inconsistent. Four studies (n = 3993/5879; 68%), reported clear associations between hypertension and ADNC. Another four suggested that antihypertensive medication may protect against ADNC. Larger studies with longer follow-up reported the strongest relationships. Our findings suggest a positive association between hypertension and ADNC, but effects may be modest, and possibly attenuate with higher hypertension age and antihypertensive medication use. Investigating interactions among plaques, tangles, cerebrovascular pathology, and dementia may be key in better understanding hypertension's role in dementia development.
Collapse
Affiliation(s)
- Herrer Abdulrahman
- Department of NeurologyAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
- Department of NeurologyRadboud University Medical CenterDonders Institute for BrainCognition and BehaviorNijmegenthe Netherlands
| | - Jan Willem van Dalen
- Department of NeurologyAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
- Department of NeurologyRadboud University Medical CenterDonders Institute for BrainCognition and BehaviorNijmegenthe Netherlands
| | - Melina den Brok
- Department of NeurologyAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
- Department of NeurologyRadboud University Medical CenterDonders Institute for BrainCognition and BehaviorNijmegenthe Netherlands
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institute SeattleSeattleWashingtonUSA
| | - Edo Richard
- Department of NeurologyRadboud University Medical CenterDonders Institute for BrainCognition and BehaviorNijmegenthe Netherlands
- Department of Public and Occupational HealthAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
34
|
Yau WYW, Shirzadi Z, Yang HS, Ikoba AP, Rabin JS, Properzi MJ, Kirn DR, Schultz AP, Rentz DM, Johnson KA, Sperling RA, Chhatwal JP. Tau Mediates Synergistic Influence of Vascular Risk and Aβ on Cognitive Decline. Ann Neurol 2022; 92:745-755. [PMID: 35880989 PMCID: PMC9650958 DOI: 10.1002/ana.26460] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Elevated vascular risk and beta-amyloid (Aβ) burden have been synergistically associated with cognitive decline in preclinical Alzheimer's disease (AD), although the underlying mechanisms remain unclear. We examined whether accelerated longitudinal tau accumulation mediates the vascular risk-Aβ interaction on cognitive decline. METHODS We included 175 cognitively unimpaired older adults (age 70.5 ± 8.0 years). Baseline vascular risk was quantified using the office-based Framingham Heart Study general cardiovascular disease risk score (FHS-CVD). Baseline Aβ burden was measured with Pittsburgh Compound-B positron emission tomography (PET). Tau burden was measured longitudinally (3.6 ± 1.5 years) with Flortaucipir PET, focusing on inferior temporal cortex (ITC). Cognition was assessed longitudinally (7.0 ± 2.0 years) using the Preclinical Alzheimer's Cognitive Composite. Linear mixed effects models examined the interactive effects of baseline vascular risk and Aβ on longitudinal ITC tau. Additionally, moderated mediation was used to determine whether tau accumulation mediated the FHS-CVD*Aβ effect on cognitive decline. RESULTS We observed a significant interaction between elevated baseline FHS-CVD and Aβ on greater ITC tau accumulation (p = 0.004), even in individuals with Aβ burden below the conventional threshold for amyloid positivity. Examining individual vascular risk factors, we found elevated systolic blood pressure and body mass index showed independent interactions with Aβ on longitudinal tau (both p < 0.0001). ITC tau accumulation mediated 33% of the interactive association of FHS-CVD and Aβ on cognitive decline. INTERPRETATION Vascular risks interact with subthreshold levels of Aβ to promote cognitive decline, partially by accelerating early neocortical tau accumulation. Our findings support vascular risk reduction, especially treating hypertension and obesity, to attenuate Aβ-related tau pathology and reduce late-life cognitive decline. ANN NEUROL 2022;92:745-755.
Collapse
Affiliation(s)
- Wai-Ying Wendy Yau
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Zahra Shirzadi
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hyun-Sik Yang
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | - Akpevweoghene P Ikoba
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Dylan R Kirn
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
35
|
Weigand AJ, Hamlin AM, Breton J, Clark AL. Cerebral blood flow, tau imaging, and memory associations in cognitively unimpaired older adults. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100153. [PMID: 36353072 PMCID: PMC9637859 DOI: 10.1016/j.cccb.2022.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Objective Cerebral blood flow (CBF) has been independently linked to cognitive impairment and traditional Alzheimer's disease (AD) pathology (e.g., amyloid-beta [Aβ], tau) in older adults. However, less is known about the possible interactive effects of CBF, Aβ, and tau on memory performance. The present study examined whether CBF moderates the effect of Aβ and tau on objective and subjective memory within cognitively unimpaired (CU) older adults. Methods Participants included 54 predominately white CU older adults from the Alzheimer's Disease Neuroimaging Initiative. Multiple linear regression models examined meta-temporal CBF associations with (1) meta-temporal tau PET adjusting for cortical Aβ PET and (2) and cortical Aβ PET adjusting for tau PET. The CBF and tau meta region was an average of 5 distinct temporal lobe regions. CBF interactions with Aβ or tau PET on memory performance were also examined. Covariates for all models included age, sex, education, pulse pressure, APOE-ε4 positivity, and imaging acquisition date differences. Results CBF was significantly negatively associated with tau PET (t = -2.16, p = .04) but not Aβ PET (t = 0.98, p = .33). Results revealed a CBF by tau PET interaction such that there was a stronger effect of tau PET on objective (t = 2.51, p = .02) and subjective (t = -2.67, p = .01) memory outcomes among individuals with lower levels of CBF. Conclusions Cerebrovascular and tau pathologies may interact to influence cognitive performance. This study highlights the need for future vascular risk interventions, which could offer a scalable and cost-effective method for AD prevention.
Collapse
Affiliation(s)
- Alexandra J. Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, United States
| | - Abbey M. Hamlin
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| | - Jordana Breton
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| | - Alexandra L. Clark
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| |
Collapse
|
36
|
Blessing EM, Parekh A, Betensky RA, Babb J, Saba N, Debure L, Varga AW, Ayappa I, Rapoport DM, Butler TA, de Leon MJ, Wisniewski T, Lopresti BJ, Osorio RS. Association between lower body temperature and increased tau pathology in cognitively normal older adults. Neurobiol Dis 2022; 171:105748. [PMID: 35550158 PMCID: PMC9751849 DOI: 10.1016/j.nbd.2022.105748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/25/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Preclinical studies suggest body temperature (Tb) and consequently brain temperature has the potential to bidirectionally interact with tau pathology in Alzheimer's Disease (AD). Tau phosphorylation is substantially increased by a small (<1 °C) decrease in temperature within the human physiological range, and thermoregulatory nuclei are affected by tau pathology early in the AD continuum. In this study we evaluated whether Tb (as a proxy for brain temperature) is cross-sectionally associated with clinically utilized markers of tau pathology in cognitively normal older adults. METHODS Tb was continuously measured with ingestible telemetry sensors for 48 h. This period included two nights of nocturnal polysomnography to delineate whether Tb during waking vs sleep is differentially associated with tau pathology. Tau phosphorylation was assessed with plasma and cerebrospinal fluid (CSF) tau phosphorylated at threonine 181 (P-tau), sampled the day following Tb measurement. In addition, neurofibrillary tangle (NFT) burden in early Braak stage regions was imaged with PET-MR using the [18F]MK-6240 radiotracer on average one month later. RESULTS Lower Tb was associated with increased NFT burden, as well as increased plasma and CSF P-tau levels (p < 0.05). NFT burden was associated with lower Tb during waking (p < 0.05) but not during sleep intervals. Plasma and CSF P-tau levels were highly correlated with each other (p < 0.05), and both variables were correlated with tau tangle radiotracer uptake (p < 0.05). CONCLUSIONS These results, the first available for human, suggest that lower Tb in older adults may be associated with increased tau pathology. Our findings add to the substantial preclinical literature associating lower body and brain temperature with tau hyperphosphorylation. CLINICAL TRIAL NUMBER NCT03053908.
Collapse
Affiliation(s)
- Esther M Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Ankit Parekh
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Rebecca A Betensky
- Department of NYU School of Global Public Health, New York, NY 10016, United States of America.
| | - James Babb
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Natalie Saba
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Ludovic Debure
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Andrew W Varga
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Indu Ayappa
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - David M Rapoport
- Mount Sinai Integrative Sleep Center, Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, United States of America.
| | - Tracy A Butler
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Mony J de Leon
- Department of Neurology, Weill Cornell Medicine, New York, NY 10065, United States of America.
| | - Thomas Wisniewski
- Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America.
| | - Ricardo S Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, United States of America; Alzheimer's Disease Research Center, Department of Neurology, NYU Grossman School of Medicine, New York, NY 10016, United States of America.
| |
Collapse
|
37
|
Ottoy J, Ozzoude M, Zukotynski K, Adamo S, Scott C, Gaudet V, Ramirez J, Swardfager W, Cogo-Moreira H, Lam B, Bhan A, Mojiri P, Kang MS, Rabin JS, Kiss A, Strother S, Bocti C, Borrie M, Chertkow H, Frayne R, Hsiung R, Laforce RJ, Noseworthy MD, Prato FS, Sahlas DJ, Smith EE, Kuo PH, Sossi V, Thiel A, Soucy JP, Tardif JC, Black SE, Goubran M. Vascular burden and cognition: Mediating roles of neurodegeneration and amyloid PET. Alzheimers Dement 2022; 19:1503-1517. [PMID: 36047604 DOI: 10.1002/alz.12750] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 11/06/2022]
Abstract
It remains unclear to what extent cerebrovascular burden relates to amyloid beta (Aβ) deposition, neurodegeneration, and cognitive dysfunction in mixed disease populations with small vessel disease and Alzheimer's disease (AD) pathology. In 120 subjects, we investigated the association of vascular burden (white matter hyperintensity [WMH] volumes) with cognition. Using mediation analyses, we tested the indirect effects of WMH on cognition via Aβ deposition (18 F-AV45 positron emission tomography [PET]) and neurodegeneration (cortical thickness or 18 F fluorodeoxyglucose PET) in AD signature regions. We observed that increased total WMH volume was associated with poorer performance in all tested cognitive domains, with the strongest effects observed for semantic fluency. These relationships were mediated mainly via cortical thinning, particularly of the temporal lobe, and to a lesser extent serially mediated via Aβ and cortical thinning of AD signature regions. WMH volumes differentially impacted cognition depending on lobar location and Aβ status. In summary, our study suggests mainly an amyloid-independent pathway in which vascular burden affects cognitive function via localized neurodegeneration. HIGHLIGHTS: Alzheimer's disease often co-exists with vascular pathology. We studied a unique cohort enriched for high white matter hyperintensities (WMH). High WMH related to cognitive impairment of semantic fluency and executive function. This relationship was mediated via temporo-parietal atrophy rather than metabolism. This relationship was, to lesser extent, serially mediated via amyloid beta and atrophy.
Collapse
Affiliation(s)
- Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Miracle Ozzoude
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Katherine Zukotynski
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Departments of Medicine and Radiology, McMaster University, Hamilton, Ontario, Canada.,Department of Medical Imaging, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sabrina Adamo
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Scott
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Vincent Gaudet
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Hugo Cogo-Moreira
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Department of Education, ICT and Learning, Østfold University College, Halden, Norway
| | - Benjamin Lam
- Department of Medicine (Division of Neurology), University of Toronto, Toronto, Ontario, Canada
| | - Aparna Bhan
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Parisa Mojiri
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer S Rabin
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Alex Kiss
- Department of Research Design and Biostatistics, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Stephen Strother
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,The Rotman Research Institute Baycrest, University of Toronto, Toronto, Ontario, Canada
| | - Christian Bocti
- Département de Médecine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Michael Borrie
- Lawson Health Research Institute, Western University, London, Ontario, Canada
| | - Howard Chertkow
- Jewish General Hospital and Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Richard Frayne
- Departments of Radiology and Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Robin Hsiung
- Physics and Astronomy Department and DM Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert Jr Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Université Laval, Quebec City, Quebec, Canada
| | - Michael D Noseworthy
- Department of Electrical and Computer Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Frank S Prato
- Lawson Health Research Institute, Western University, London, Ontario, Canada
| | | | - Eric E Smith
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Phillip H Kuo
- Department of Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | - Vesna Sossi
- Physics and Astronomy Department and DM Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexander Thiel
- Jewish General Hospital and Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Sandra E Black
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine (Division of Neurology), University of Toronto, Toronto, Ontario, Canada.,Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Maged Goubran
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
38
|
Lao PJ, Boehme AK, Morales C, Laing KK, Chesebro A, Igwe KC, Gutierrez J, Gu Y, Stern Y, Schupf N, Manly JJ, Mayeux R, Brickman AM. Amyloid, cerebrovascular disease, and neurodegeneration biomarkers are associated with cognitive trajectories in a racially and ethnically diverse, community-based sample. Neurobiol Aging 2022; 117:83-96. [PMID: 35679806 PMCID: PMC9997572 DOI: 10.1016/j.neurobiolaging.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023]
Abstract
We characterized the additive contribution of cerebrovascular biomarkers to amyloid and neurodegeneration biomarkers (AV(N)) when modeling prospective, longitudinal cognitive trajectories within 3 major racial/ethnic groups. Participants (n = 172; age = 69-96 years; 62% women; 31%/49%/20% Non-Hispanic White/Non-Hispanic Black/Hispanic) from the Washington Heights-Inwood Columbia Aging Project were assessed for amyloid (Florbetaben PET), neurodegeneration (cortical thickness, hippocampal volume), and cerebrovascular disease (white matter hyperintensity (WMH), infarcts). Neuropsychological assessments occurred every 2.3 ± 0.6 years for up to 6 visits (follow-up time: 4.2 ± 3.2 years). Linear mixed-effects models were stratified by race/ethnicity groups. Higher amyloid was associated with faster memory decline in all 3 racial/ethnic groups, but was related to faster cognitive decline beyond memory in minoritized racial/ethnic groups. Higher WMH was associated with faster language, processing speed/executive function, and visuospatial ability decline in Non-Hispanic Black participants, while infarcts were associated with faster processing speed/executive function decline in Non-Hispanic White participants. Complementary information from AD, neurodegenerative, and cerebrovascular biomarkers explain decline in multiple cognitive domains, which may differ within each racial/ethnic group. Importantly, treatment strategies exist to minimize vascular contributions to cognitive decline.
Collapse
Affiliation(s)
- Patrick J Lao
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Amelia K Boehme
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Clarissa Morales
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Krystal K Laing
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Anthony Chesebro
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Kay C Igwe
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jose Gutierrez
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Yian Gu
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Yaakov Stern
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Nicole Schupf
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jennifer J Manly
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Richard Mayeux
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, USA; Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Adam M Brickman
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Gertrude H. Sergievsky, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
39
|
Rabin JS, Nichols E, La Joie R, Casaletto KB, Palta P, Dams-O’Connor K, Kumar RG, George KM, Satizabal CL, Schneider JA, Pa J, Brickman AM. Cerebral amyloid angiopathy interacts with neuritic amyloid plaques to promote tau and cognitive decline. Brain 2022; 145:2823-2833. [PMID: 35759327 PMCID: PMC9420012 DOI: 10.1093/brain/awac178] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
Accumulating data suggest that cerebrovascular disease contributes to Alzheimer's disease pathophysiology and progression toward dementia. Cerebral amyloid angiopathy is a form of cerebrovascular pathology that results from the build-up of β-amyloid in the vessel walls. Cerebral amyloid angiopathy commonly co-occurs with Alzheimer's disease pathology in the ageing brain and increases the risk of Alzheimer's disease dementia. In the present study, we examined whether cerebral amyloid angiopathy influences tau deposition and cognitive decline independently or synergistically with parenchymal β-amyloid burden. Secondly, we examined whether tau burden mediates the association between cerebral amyloid angiopathy and cognitive decline. We included data from autopsied subjects recruited from one of three longitudinal clinical-pathological cohort studies: the Rush Memory and Aging Project, the Religious Orders Study and the Minority Aging Research Study. Participants completed annual clinical and cognitive evaluations and underwent brain autopsy. Cerebral amyloid angiopathy pathology was rated as none, mild, moderate or severe. Bielschowsky silver stain was used to visualize neuritic β-amyloid plaques and neurofibrillary tangles. We used linear regression and linear mixed models to test independent versus interactive associations of cerebral amyloid angiopathy and neuritic plaque burden with tau burden and longitudinal cognitive decline, respectively. We used causal mediation models to examine whether tau mediates the association between cerebral amyloid angiopathy and cognitive decline. The study sample included 1722 autopsied subjects (age at baseline = 80.2 ± 7.1 years; age at death = 89.5 ± 6.7 years; 68% females). Cerebral amyloid angiopathy interacted with neuritic plaques to accelerate tau burden and cognitive decline. Specifically, those with more severe cerebral amyloid angiopathy pathology and higher levels of neuritic plaque burden had greater tau burden and faster cognitive decline. We also found that tau mediated the association between cerebral amyloid angiopathy and cognitive decline among participants with higher neuritic plaque burden. In summary, more severe levels of cerebral amyloid angiopathy and higher parenchymal β-amyloid burden interacted to promote cognitive decline indirectly via tau deposition. These results highlight the dynamic interplay between cerebral amyloid angiopathy and Alzheimer's disease pathology in accelerating progression toward dementia. These findings have implications for Alzheimer's disease clinical trials and therapeutic development.
Collapse
Affiliation(s)
- Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences
Centre, University of Toronto, Toronto, Canada M4N 3M5
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program,
Sunnybrook Research Institute, Toronto, Ontario,
Canada M4N 3M5
- Rehabilitation Sciences Institute, University of Toronto,
Toronto, Canada M5G 1V7
| | - Emma Nichols
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public
Health, Baltimore, MD, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for
Neurosciences, University of California, San Francisco,
CA, USA 94158
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for
Neurosciences, University of California, San Francisco,
CA, USA 94158
| | - Priya Palta
- Departments of Medicine and Epidemiology, Columbia University Irving
Medical Center, New York, NY, USA
| | - Kristen Dams-O’Connor
- Department of Rehabilitation and Human Performance, Icahn School of
Medicine at Mount Sinai, New York, NY, USA 10029
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, NY, USA 10029
| | - Raj G Kumar
- Department of Rehabilitation and Human Performance, Icahn School of
Medicine at Mount Sinai, New York, NY, USA 10029
| | - Kristen M George
- Department of Public Health Sciences, University of California Davis School
of Medicine, Davis, CA, USA
| | - Claudia L Satizabal
- Department of Population Health Science and Biggs Institute for Alzheimer’s
and Neurodegenerative Diseases, UT Health San Antonio, San
Antonio, TX, USA
- Department of Neurology, Boston University School of
Medicine, Boston, MA, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical
Center, Chicago, IL, USA
| | - Judy Pa
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Department of
Neurology, University of Southern California, Los Angeles,
CA, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain,
Department of Neurology, College of Physicians and Surgeons, Columbia
University, New York, NY, USA 10032
| |
Collapse
|
40
|
Jullienne A, Quan R, Szu JI, Trinh MV, Behringer EJ, Obenaus A. Progressive Vascular Abnormalities in the Aging 3xTg-AD Mouse Model of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081967. [PMID: 36009514 PMCID: PMC9405684 DOI: 10.3390/biomedicines10081967] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/01/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular dysfunction and structural abnormalities in Alzheimer’s disease (AD) are known to contribute to the progression of the pathology, and studies have tended to ignore the role of the vasculature in AD progression. We utilized the 3xTg-AD mouse model of AD to examine individual cerebral vessels and the cortical vascular network across the lifespan. Our vessel painting approach was used to label the entire cortical vasculature, followed by epifluorescence microscopy. The middle cerebral artery (MCA) tree was assessed with confocal microscopy, and a new method was developed to assess branching patterns as a measure of aging-related changes. We found that vascular remodeling was profoundly altered at 4–6 months of age, when the 3xTg-AD mouse is known to transition to cognitive impairment and Aβ deposition in both sexes. Analysis of vascular features (density, junctions, length) of the MCA territory highlighted sex-dependent differences across the 3xTg-AD mouse lifespan, with no alterations in branching patterns. Our current cerebrovascular angioarchitectural analyses demonstrate progressive alterations in individual cortical vessels, as well as in the vascular network of the cortex. These new findings advance our understanding of brain anatomy and physiology in the 3xTg-AD mouse, while potentially identifying unique diagnostic signatures of AD progression.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Ryan Quan
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Jenny I. Szu
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Michelle V. Trinh
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Erik J. Behringer
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, CA 92697, USA
- Department of Basic Sciences, Loma Linda University, Loma Linda, CA 92350, USA
- Correspondence:
| |
Collapse
|
41
|
White Matter, Behavioral Neurology, and the Influence of Corticocentrism. Cogn Behav Neurol 2022; 35:147-152. [PMID: 35486535 DOI: 10.1097/wnn.0000000000000302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/26/2022]
Abstract
White matter in the human brain occupies roughly the same volume as gray matter but has received far less attention in behavioral neurology and related disciplines. In particular, the cerebral cortex has long dominated thinking about the organization of brain-behavior relationships. As a result, subcortical structures, including deep gray matter and, most notably, white matter, have been accorded relatively little neuroscientific study compared with the extensive work devoted to the cerebral cortex. The influence of corticocentrism can be explained by several factors, including historical precedent in neurology strongly emphasizing the importance of the cortex, a preponderance of investigative methods that selectively target this structure, and a misinterpretation of comparative neuroanatomic data gathered from normal brains. This paper will describe the background of the corticocentric bias and emphasize that white matter merits its own place within the study of the higher functions. Although corticocentrism continues to exert a powerful impact on behavioral neurology, considerable progress is being made in the study of white matter-a development that promises to expand our knowledge of the normal brain and lead to an improved understanding of how it mediates behavior. In turn, a range of vexing neurologic and psychiatric disorders may become better illuminated by considering pathology within, or dysfunction of, white matter tracts. A complete appreciation of brain-behavior relationships requires an understanding not only of the outermost layer of the cerebral hemispheres, but also of white matter connectivity that links gray matter regions into distributed neural networks that subserve cognition and emotion.
Collapse
|
42
|
Veitch DP, Weiner MW, Aisen PS, Beckett LA, DeCarli C, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Okonkwo O, Perrin RJ, Petersen RC, Rivera‐Mindt M, Saykin AJ, Shaw LM, Toga AW, Tosun D, Trojanowski JQ. Using the Alzheimer's Disease Neuroimaging Initiative to improve early detection, diagnosis, and treatment of Alzheimer's disease. Alzheimers Dement 2022; 18:824-857. [PMID: 34581485 PMCID: PMC9158456 DOI: 10.1002/alz.12422] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The Alzheimer's Disease Neuroimaging Initiative (ADNI) has accumulated 15 years of clinical, neuroimaging, cognitive, biofluid biomarker and genetic data, and biofluid samples available to researchers, resulting in more than 3500 publications. This review covers studies from 2018 to 2020. METHODS We identified 1442 publications using ADNI data by conventional search methods and selected impactful studies for inclusion. RESULTS Disease progression studies supported pivotal roles for regional amyloid beta (Aβ) and tau deposition, and identified underlying genetic contributions to Alzheimer's disease (AD). Vascular disease, immune response, inflammation, resilience, and sex modulated disease course. Biologically coherent subgroups were identified at all clinical stages. Practical algorithms and methodological changes improved determination of Aβ status. Plasma Aβ, phosphorylated tau181, and neurofilament light were promising noninvasive biomarkers. Prognostic and diagnostic models were externally validated in ADNI but studies are limited by lack of ethnocultural cohort diversity. DISCUSSION ADNI has had a profound impact in improving clinical trials for AD.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of RadiologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of PsychiatryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Laurel A. Beckett
- Division of Biostatistics, Department of Public Health SciencesUniversity of California DavisDavisCaliforniaUSA
| | - Charles DeCarli
- Department of Neurology and Center for NeuroscienceUniversity of California DavisDavisCaliforniaUSA
| | - Robert C. Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Broad Institute, Ariadne Labsand Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health SciencesUniversity of California DavisDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | | | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences and Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuroimaging, USC Stevens Institute of Neuroimaging and Informatics, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of RadiologyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | |
Collapse
|
43
|
Kapasi A, Yu L, Petyuk V, Arfanakis K, Bennett DA, Schneider JA. Association of small vessel disease with tau pathology. Acta Neuropathol 2022; 143:349-362. [PMID: 35044500 PMCID: PMC8858293 DOI: 10.1007/s00401-021-02397-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Emerging evidence suggests that small vessel disease (SVD) is a risk factor for clinical dementia and may contribute to AD neuropathological changes. Watershed brain regions are located at the most distal areas between arterial territories, making them vulnerable to SVD-related changes. We examined the association of pathologic markers of SVD, specifically arteriolosclerosis in watershed brain regions, with AD pathologic changes. Participants (N = 982; mean age-at-death = 90; 69% women) were enrolled as part of one of two cohort studies of aging and dementia. At autopsy, neuropathological evaluation included semi-quantitative grading of arteriolosclerosis pathology from 2 cortical watershed regions: the anterior watershed (AWS) and posterior watershed (PWS), densities for cortical β-amyloid and tau-tangle pathology, and other common age-related pathologies. Linear regression models examined the association of watershed arteriolosclerosis pathology with β-amyloid and tau-tangle burden. In follow-up analyses, available ex-vivo MRI and proteomics data in a subset of decedents were leveraged to examine the association of whole brain measure of WMH, as a presumed MRI marker of SVD, with β-amyloid and tau-tangle burden, as well as to examine the association of watershed arteriolosclerosis with proteomic tau. Watershed arteriolosclerosis was common, with 45% of older persons having moderate-to-severe arteriolosclerosis pathology in the AWS region, and 35% in the PWS. In fully adjusted models that controlled for demographics and common age-related pathologies, an increase in severity of PWS arteriolosclerosis was associated with a higher burden of tau-tangle burden, specifically neocortical tau burden, but not with β-amyloid. AWS arteriolosclerosis was not associated with β-amyloid or tau pathology. Ex-vivo WMH was associated with greater tau-tangle pathology burden but not β-amyloid. Furthermore, PWS arteriolosclerosis was associated with higher abundance of tau phosphopeptides, that promote formation of tau aggregates. These data provide compelling evidence that SVD, specifically posterior watershed arteriolosclerosis pathology, is linked with tau pathological changes in the aging brain.
Collapse
Affiliation(s)
- Alifiya Kapasi
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Chicago, IL, 60612, USA.
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA.
| | - L Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - V Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - K Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Chicago, IL, 60612, USA
- Department of Diagnostic Radiology and Nuclear Medicine, Chicago, IL, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - D A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - J A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison Street, Chicago, IL, 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
44
|
Kochsiek J, O’Donnell LJ, Zhang F, Bonke EM, Sollmann N, Tripodis Y, Wiegand TLT, Kaufmann D, Umminger L, Di Biase MA, Kaufmann E, Schultz V, Alosco ML, Martin BM, Lin AP, Coleman MJ, Rathi Y, Pasternak O, Bouix S, Stern RA, Shenton ME, Koerte IK. Exposure to Repetitive Head Impacts Is Associated With Corpus Callosum Microstructure and Plasma Total Tau in Former Professional American Football Players. J Magn Reson Imaging 2021; 54:1819-1829. [PMID: 34137112 PMCID: PMC8633029 DOI: 10.1002/jmri.27774] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Exposure to repetitive head impacts (RHI) is associated with an increased risk of later-life neurobehavioral dysregulation and neurodegenerative disease. The underlying pathomechanisms are largely unknown. PURPOSE To investigate whether RHI exposure is associated with later-life corpus callosum (CC) microstructure and whether CC microstructure is associated with plasma total tau and neuropsychological/neuropsychiatric functioning. STUDY TYPE Retrospective cohort study. POPULATION Seventy-five former professional American football players (age 55.2 ± 8.0 years) with cognitive, behavioral, and mood symptoms. FIELD STRENGTH/SEQUENCE Diffusion-weighted echo-planar MRI at 3 T. ASSESSMENT Subjects underwent diffusion MRI, venous puncture, neuropsychological testing, and completed self-report measures of neurobehavioral dysregulation. RHI exposure was assessed using the Cumulative Head Impact Index (CHII). Diffusion MRI measures of CC microstructure (i.e., free-water corrected fractional anisotropy (FA), trace, radial diffusivity (RD), and axial diffusivity (AD)) were extracted from seven segments of the CC (CC1-7), using a tractography clustering algorithm. Neuropsychological tests were selected: Trail Making Test Part A (TMT-A) and Part B (TMT-B), Controlled Oral Word Association Test (COWAT), Stroop Interference Test, and the Behavioral Regulation Index (BRI) from the Behavior Rating Inventory of Executive Function, Adult version (BRIEF-A). STATISTICAL TESTS Diffusion MRI metrics were tested for associations with RHI exposure, plasma total tau, neuropsychological performance, and neurobehavioral dysregulation using generalized linear models for repeated measures. RESULTS RHI exposure was associated with increased AD of CC1 (correlation coefficient (r) = 0.32, P < 0.05) and with increased plasma total tau (r = 0.34, P < 0.05). AD of the anterior CC1 was associated with increased plasma total tau (CC1: r = 0.30, P < 0.05; CC2: r = 0.29, P < 0.05). Higher trace, AD, and RD of CC1 were associated with better performance (P < 0.05) in TMT-A (trace, r = 0.33; AD, r = 0.31; and RD, r = 0.28) and TMT-B (trace, r = 0.31; RD, r = 0.34). Higher FA and AD of CC2 were associated with better performance (P < 0.05) in TMT-A (FA, r = 0.36; AD, r = 0.28), TMT-B (FA, r = 0.36; AD, r = 0.27), COWAT (FA, r = 0.36; AD, r = 0.32), and BRI (AD, r = 0.29). DATA CONCLUSION These results suggest an association among RHI exposure, CC microstructure, plasma total tau, and clinical functioning in former professional American football players. LEVEL OF EVIDENCE 3 Technical Efficacy Stage: 1.
Collapse
Affiliation(s)
- Janna Kochsiek
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Lauren J. O’Donnell
- Laboratory of Mathematics in Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fan Zhang
- Laboratory of Mathematics in Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena M. Bonke
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nico Sollmann
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Alzheimer’s Disease and CTE Centers, Boston University School of Medicine, Boston, MA, USA
| | - Tim L. T. Wiegand
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - David Kaufmann
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Radiology, Charité Universitätsmedizin, Berlin, Germany
- Department of Radiology, University Hospital Augsburg, University of Augsburg, Germany
| | - Lisa Umminger
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Maria A. Di Biase
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elisabeth Kaufmann
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Vivian Schultz
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael L. Alosco
- Alzheimer’s Disease and CTE Centers, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boszon University School of Medicine, Boston, MA, USA
| | - Brett M. Martin
- Data Coordinating Center, Boston University School of Public Health, Boston, MA, USA
| | - Alexander P. Lin
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J. Coleman
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Yogesh Rathi
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ofer Pasternak
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sylvain Bouix
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert A. Stern
- Alzheimer’s Disease and CTE Centers, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boszon University School of Medicine, Boston, MA, USA
- Departments of Neurosugery and Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Martha E. Shenton
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Inga K. Koerte
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Chum PP, Hakim MA, Behringer EJ. Cerebrovascular microRNA Expression Profile During Early Development of Alzheimer's Disease in a Mouse Model. J Alzheimers Dis 2021; 85:91-113. [PMID: 34776451 DOI: 10.3233/jad-215223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Emerging evidence demonstrates association of Alzheimer's disease (AD) with impaired delivery of blood oxygen and nutrients to and throughout the brain. The cerebral circulation plays multiple roles underscoring optimal brain perfusion and cognition entailing moment-to-moment blood flow control, vascular permeability, and angiogenesis. With currently no effective treatment to prevent or delay the progression of AD, cerebrovascular microRNA (miRNA) markers corresponding to post-transcriptional regulation may distinguish phases of AD. OBJECTIVE We tested the hypothesis that cerebrovascular miRNA expression profiles indicate developmental stages of AD pathology. METHODS Total RNA was isolated from total brain vessel segments of male and female 3xTg-AD mice [young, 1-2 mo; cognitive impairment (CI), 4-5 mo; extracellular amyloid-β plaques (Aβ), 6-8 mo; plaques+neurofibrillary tangles (AβT), 12-15 mo]. NanoString technology nCounter miRNA Expression panel for mouse was used to screen for 599 miRNAs. RESULTS Significant (p < 0.05) downregulation of various miRNAs indicated transitions from young to CI (e.g., let-7g & miR-1944, males; miR-133a & miR-2140, females) and CI to Aβ (e.g., miR-99a, males) but not from Aβ to AβT. In addition, altered expression of select miRNAs from overall Pre-AD (young + CI) versus AD (Aβ+ AβT) were detected in both males (let-7d, let-7i, miR-23a, miR-34b-3p, miR-99a, miR-126-3p, miR-132, miR-150, miR-151-5p, miR-181a) and females (miR-150, miR-539). Altogether, at least 20 cerebrovascular miRNAs effectively delineate AD versus Pre-AD pathology. CONCLUSION Using the 3xTg-AD mouse model, these data demonstrate that cerebrovascular miRNAs pertaining to endothelial function, vascular permeability, angiogenesis, inflammation, and Aβ/tau metabolism can track early development of AD.
Collapse
Affiliation(s)
- Phoebe P Chum
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Md A Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | |
Collapse
|
46
|
Guo T, Landau SM, Jagust WJ. Age, vascular disease, and Alzheimer's disease pathologies in amyloid negative elderly adults. ALZHEIMERS RESEARCH & THERAPY 2021; 13:174. [PMID: 34654465 PMCID: PMC8520216 DOI: 10.1186/s13195-021-00913-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/03/2021] [Indexed: 02/04/2023]
Abstract
Background We recently reported that CSF phosphorylated tau (p-Tau181) relative to Aβ40 (CSF p-Tau/Aβ40 ratio) was less noisy and increased associations with Alzheimer’s disease (AD) biomarkers compared to CSF p-Tau181 alone. While elevations of CSF p-Tau/Aβ40 can occur in amyloid-β (Aβ) negative (Aβ-) individuals, the factors associated with these elevations and their role in neurodegeneration and cognitive decline are unknown. We aim to explore factors associated with elevated tau in CSF, and how these elevated tau are related to neurodegeneration and cognitive decline in the absence of Aβ positivity. Methods We examined relationships between CSF p-Tau/Aβ40, and CSF Aβ42/Aβ40, Aβ PET, and white matter hyperintensities (WMH) as well as vascular risk factors in 149 cognitively unimpaired and 52 impaired individuals who were presumably not on the Alzheimer’s disease (AD) pathway due to negative Aβ status on both CSF and PET. Subgroups had 18F-fluorodeoxyglucose (FDG) PET and adjusted hippocampal volume (aHCV), and longitudinal measures of CSF, aHCV, FDG PET, and cognition data, so we examined CSF p-Tau/Aβ40 associations with these measures as well. Results Elevated CSF p-Tau/Aβ40 was associated with older age, male sex, greater WMH, and hypertension as well as a pattern of hippocampal atrophy and temporoparietal hypometabolism characteristic of AD. Lower CSF Aβ42/Aβ40, higher WMH, and hypertension but not age, sex, Aβ PET, APOE-ε4 status, body mass index, smoking, and hyperlipidemia at baseline predicted CSF p-Tau/Aβ40 increases over approximately 5 years of follow-up. The relationship between CSF p-Tau/Aβ40 and subsequent cognitive decline was partially or fully explained by neurodegenerative measurements. Conclusions These data provide surprising clues as to the etiology and significance of tau pathology in the absence of Aβ. It seems likely that, in addition to age, both cerebrovascular disease and subthreshold levels of Aβ are related to this tau accumulation. Crucially, this phenotype of CSF tau elevation in amyloid-negative individuals share features with AD such as a pattern of metabolic decline and regional brain atrophy. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00913-5.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | | |
Collapse
|
47
|
Grimm A. Impairments in Brain Bioenergetics in Aging and Tau Pathology: A Chicken and Egg Situation? Cells 2021; 10:2531. [PMID: 34685510 PMCID: PMC8533761 DOI: 10.3390/cells10102531] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
The brain is the most energy-consuming organ of the body and impairments in brain energy metabolism will affect neuronal functionality and viability. Brain aging is marked by defects in energetic metabolism. Abnormal tau protein is a hallmark of tauopathies, including Alzheimer's disease (AD). Pathological tau was shown to induce bioenergetic impairments by affecting mitochondrial function. Although it is now clear that mutations in the tau-coding gene lead to tau pathology, the causes of abnormal tau phosphorylation and aggregation in non-familial tauopathies, such as sporadic AD, remain elusive. Strikingly, both tau pathology and brain hypometabolism correlate with cognitive impairments in AD. The aim of this review is to discuss the link between age-related decrease in brain metabolism and tau pathology. In particular, the following points will be discussed: (i) the common bioenergetic features observed during brain aging and tauopathies; (ii) how age-related bioenergetic defects affect tau pathology; (iii) the influence of lifestyle factors known to modulate brain bioenergetics on tau pathology. The findings compiled here suggest that age-related bioenergetic defects may trigger abnormal tau phosphorylation/aggregation and cognitive impairments after passing a pathological threshold. Understanding the effects of aging on brain metabolism may therefore help to identify disease-modifying strategies against tau-induced neurodegeneration.
Collapse
Affiliation(s)
- Amandine Grimm
- Transfaculty Research Platform Molecular and Cognitive Neuroscience, University of Basel, 4002 Basel, Switzerland;
- Neurobiology Lab for Brain Aging and Mental Health, Psychiatric University Clinics, 4002 Basel, Switzerland
- Life Sciences Training Facility, University of Basel, 4055 Basel, Switzerland
| |
Collapse
|
48
|
Li S, Wang C, Wang Z, Tan J. Involvement of cerebrovascular abnormalities in the pathogenesis and progression of Alzheimer's disease: an adrenergic approach. Aging (Albany NY) 2021; 13:21791-21806. [PMID: 34479211 PMCID: PMC8457611 DOI: 10.18632/aging.203482] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/17/2021] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD), as the most common neurodegenerative disease in elder population, is pathologically characterized by β-amyloid (Aβ) plaques, neurofibrillary tangles composed of highly-phosphorylated tau protein and consequently progressive neurodegeneration. However, both Aβ and tau fails to cover the whole pathological process of AD, and most of the Aβ- or tau-based therapeutic strategies are all failed. Increasing lines of evidence from both clinical and preclinical studies have indicated that age-related cerebrovascular dysfunctions, including the changes in cerebrovascular microstructure, blood-brain barrier integrity, cerebrovascular reactivity and cerebral blood flow, accompany or even precede the development of AD-like pathologies. These findings may raise the possibility that cerebrovascular changes are likely pathogenic contributors to the onset and progression of AD. In this review, we provide an appraisal of the cerebrovascular alterations in AD and the relationship to cognitive impairment and AD pathologies. Moreover, the adrenergic mechanisms leading to cerebrovascular and AD pathologies were further discussed. The contributions of early cerebrovascular factors, especially through adrenergic mechanisms, should be considered and treasured in the diagnostic, preventative, and therapeutic approaches to address AD.
Collapse
Affiliation(s)
- Song Li
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Che Wang
- Department of Pharmaceutical Chemistry, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
49
|
Abstract
UNLABELLED Exercise is associated with higher cognitive function and is a promising intervention to reduce the risk of dementia. With advancing age, there are changes in the vasculature that have important clinical implications for brain health and cognition. Primary aging and vascular risk factors are associated with increases in arterial stiffness and pulse pressure, and reductions in peripheral vascular function. OBJECTIVE The purpose is to discuss the epidemiological, observational, and mechanistic evidence regarding the link between age-related changes in vascular health and brain health. METHODS We performed a literature review and integrated with our published data. RESULTS Epidemiological evidence suggests a link between age-related increases in arterial stiffness and lower cognitive function, which may be mediated by cerebral vascular function, including cerebral vasoreactivity and cerebral pulsatility. Age-associated impairments in central arterial stiffness and peripheral vascular function have been attenuated or reversed through lifestyle behaviors such as exercise. Greater volumes of habitual exercise and higher cardiorespiratory fitness are associated with beneficial effects on both peripheral vascular health and cognition. Yet, the extent to which exercise directly influences cerebral vascular function and brain health, as well as the associated mechanisms remains unclear. CONCLUSION Although there is evidence that exercise positively impacts cerebral vascular function, more research is necessary in humans to optimize experimental protocols and address methodological limitations and physiological considerations. Understanding the impact of exercise on cerebral vascular function is important for understanding the association between exercise and brain health and may inform future intervention studies that seek to improve cognition.
Collapse
|
50
|
Zhang S, Kwapong WR, Yang T, Liu P, Tuo Q, Cheng Y, Li X, Liu M, Lei P, Wu B. Choriocapillaris Changes Are Correlated With Disease Duration and MoCA Score in Early-Onset Dementia. Front Aging Neurosci 2021; 13:656750. [PMID: 33927609 PMCID: PMC8076507 DOI: 10.3389/fnagi.2021.656750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/24/2021] [Indexed: 02/05/2023] Open
Abstract
Purpose: Imaging of the choroid may detect the microvascular changes associated with early-onset dementia (EOD) and may represent an indicator for detection of the disease. We aimed to analyze the in vivo choriocapillaris (CC) flow density in EOD patients using optical coherence tomography angiography (OCTA) and evaluate the association with its clinical measures. Methods: This cross-sectional study used the OCTA to image and analyze the choriocapillaris (CC) of 25 EOD patients and 20 healthy controls. Choriocapillaris flow density in the 3 mm area and 6 mm area was measured by an inbuilt algorithm in the OCT tool. Brain volume using magnetic resonance imaging and cognitive assessment was done and recorded. Results: Significantly reduced capillary flow density of the choriocapillaris was seen in EOD patients when compared to healthy controls in the 3.0 mm (P = 0.001) and 6.0 mm (P < 0.001) area respectively. Montreal Cognitive Assessment (MoCA) scores in EOD patients positively correlated with choriocapillaris flow density in the 3 mm area (Rho = 0.466, P = 0.021). Disease duration of EOD patients also negatively correlated with choriocapillaris density in the 3 mm area (Rho = -0.497, P = 0.008). Discussion: Our report suggests that choriocapillaris damage may be a potential indicator of early-onset dementia. Microvascular impairment may be involved in the early phase of dementia without aging playing a role in its impairment. Clinical Trial Registration: www.ClinicalTrials.gov, ChiCTR2000041386.
Collapse
Affiliation(s)
- Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Tang Yang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Liu
- Department of Emergency, West China Hospital, Sichuan University, Chengdu, China
| | - Qingzhang Tuo
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yajun Cheng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lei
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Wu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|