1
|
Grasso SM, Santos-Santos MÁ, Clark AL. On the complexity of biomarker-driven diagnoses of Alzheimer's disease. J Neuropsychol 2025. [PMID: 40317662 DOI: 10.1111/jnp.12428] [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: 01/13/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
Updated criteria pertaining to the diagnosis of Alzheimer's disease (AD) have sparked debate over the reliance on biomarkers-particularly amyloid-β and phosphorylated tau. While biomarkers promise earlier detection and standardized criteria, the potential extension and interpretation of their use in asymptomatic individuals remains controversial. Many individuals with abnormal biomarker profiles never experience cognitive decline, raising concerns about overdiagnosis, unintended negative psychosocial consequences and the blurring line between risk and definitive diagnosis. We, and others, argue that biomarker positivity should be reframed not as a definitive diagnosis but rather as an indicator of elevated risk, particularly in the absence of cognitive symptoms. Doing so better aligns with current evidence, preserves clarity in diagnosis, and avoids unintended psychosocial consequences. Crucially, the role of cognitive reserve-influenced by education, other life experiences and structural inequities-must be considered, particularly among racially and ethnically diverse populations historically underrepresented in AD research. Biomarker thresholds (as well as neuropsychological tools) derived from predominantly non-Hispanic white cohorts may not generalize across groups, risking misclassification and inequity. As the field moves towards precision medicine and AI-driven risk models, inclusive data and culturally valid frameworks are essential. Ultimately, embracing a risk-based, multifactorial approach respects the complexity of AD and promotes equitable care. This perspective calls for interdisciplinary collaboration to refine diagnostic strategies that are scientifically grounded, socially conscious and responsive to the lived realities of diverse populations. Only then can we responsibly integrate biomarkers into practice without sacrificing nuance.
Collapse
Affiliation(s)
- Stephanie M Grasso
- Department of Speech, Language and Hearing Sciences, The University of Texas at Austin, Austin, Texas, USA
| | | | | |
Collapse
|
2
|
He J, Zhao C, Zhong S, Ouyang N, Sun G, Qiao L, Yang R, Zhao C, Liu H, Teng W, Liu X, Wang C, Liu S, Chen CS, Williamson JD, Sun Y. Blood pressure reduction and all-cause dementia in people with uncontrolled hypertension: an open-label, blinded-endpoint, cluster-randomized trial. Nat Med 2025:10.1038/s41591-025-03616-8. [PMID: 40258956 DOI: 10.1038/s41591-025-03616-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 02/26/2025] [Indexed: 04/23/2025]
Abstract
Dementia is a leading cause of death and disability worldwide. Here we tested the effectiveness of blood pressure (BP) reduction on the risk of all-cause dementia among 33,995 individuals aged ≥40 years with uncontrolled hypertension in rural China. We randomly assigned 163 villages to a non-physician community healthcare provider-led intervention and 163 villages to usual care. In the intervention group, trained non-physician community healthcare providers initiated and titrated antihypertensive medications according to a simple stepped-care protocol to achieve a systolic BP goal of <130 mm Hg and a diastolic BP goal of <80 mm Hg, with supervision from primary care physicians. Over 48 months, the net reduction in systolic BP was 22.0 mm Hg (95% confidence interval (CI) 20.6 to 23.4; P < 0.0001) and that in diastolic BP was 9.3 mm Hg (95% CI 8.7 to 10.0; P < 0.0001) in the intervention group compared to usual care. The primary outcome of all-cause dementia was significantly lower in the intervention group than in the usual care group (risk ratio: 0.85; 95% CI 0.76 to 0.95; P = 0.0035). Additionally, serious adverse events occurred less frequently in the intervention group (risk ratio: 0.94; 95% CI 0.91 to 0.98; P = 0.0006). This cluster-randomized trial indicates that intensive BP reduction is effective in lowering the risk of all-cause dementia in patients with hypertension. ClinicalTrials.gov: NCT03527719 .
Collapse
Affiliation(s)
- Jiang He
- Department of Epidemiology, Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Chuansheng Zhao
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhong
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Nanxiang Ouyang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Guozhe Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Lixia Qiao
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | | | - Chunxia Zhao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Huayan Liu
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Weiyu Teng
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Xu Liu
- Department of Neurology, First Hospital of China Medical University, Shenyang, China
| | - Chang Wang
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Songyue Liu
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China
| | - Chung-Shiuan Chen
- Tulane University Translational Science Institute, New Orleans, LA, USA
| | - Jeff D Williamson
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Section on Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Dugan R, Ramakrishnapillai S, Amant JS, Murray K, McKlveen K, Naseri M, Madden K, Bazzano L, Carmichael O. Lifespan cardiometabolic exposures are associated with midlife white matter microstructure: The Bogalusa Heart Study. Neuroimage 2025; 307:121034. [PMID: 39826773 DOI: 10.1016/j.neuroimage.2025.121034] [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: 10/25/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Compartment model analysis of diffusion MRI data provides unique information on the microstructural properties of white matter. However, studies relating compartment model microstructural measures to longitudinal cardiometabolic health data are rare. METHODS 130 cognitively healthy participants in the Bogalusa Heart Study completed diffusion MRI scans. Compartment model analysis was performed, and summary metrics were measured in organized and diffuse white matter. Multiple linear regression models were used to relate the white matter microstructure metrics to demographics and cardiometabolic risk factors. RESULTS In both organized and diffuse white matter, age was associated with worse diffusion metrics, women had better diffusion metrics than men, and African American participants had worse diffusion metrics compared to White participants. Greater blood pressure in pre-adulthood was associated with worse diffusion metrics in midlife. DISCUSSION Summary metrics from compartment model analyses of diffusion MRI data were associated cardiometabolic risk factors from youth to midlife as well as demographic factors.
Collapse
Affiliation(s)
- Reagan Dugan
- Department of Physics & Astronomy, Louisiana State University, 202 Nicholson Hall, Baton Rouge, LA, 70803, USA; Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.
| | - Sreekrishna Ramakrishnapillai
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St suite b-400, Pittsburgh, PA, 15213, USA
| | - Julia St Amant
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kori Murray
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kevin McKlveen
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Maryam Naseri
- Department of Physics & Astronomy, Louisiana State University, 202 Nicholson Hall, Baton Rouge, LA, 70803, USA; Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kaitlyn Madden
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Suite 2000, New Orleans, LA, 70112, USA
| | - Owen Carmichael
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| |
Collapse
|
4
|
Rudolph MD, Sutphen CL, Register TC, Lockhart SN, Rundle MM, Hughes TM, Bateman JR, Solingapuram Sai KK, Whitlow CT, Craft S, Mielke MM. Evaluation of plasma p-tau217 for detecting amyloid pathology in a diverse and heterogeneous community-based cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.20.25320851. [PMID: 39974029 PMCID: PMC11838994 DOI: 10.1101/2025.01.20.25320851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Studies suggest excellent performance of plasma p-tau217 for detecting amyloid pathology, though studies in more diverse populations are needed to validate previously determined cutpoints. METHODS Plasma p-tau217 utility for detecting amyloid pathology (Aβ) via amyloid PET (n=598) and/or cerebrospinal fluid (CSF; n=154) was assessed in a heterogeneous, community-based cohort in the Wake Forest Alzheimer's Disease Research Center (WFADRC). Participants (n=598) were 21% Black; 313 cognitive unimpaired (CU), 214 mild cognitive impairment (MCI), and 64 dementia (DEM); 49% prediabetic, 44% hypertensive; 29% overweight/obese; and 64% with mild-to-moderate kidney disease. Gaussian-mixture models, logistic regression, and receiver operating curve analyses were performed. RESULTS Plasma p-tau217 was associated with elevated Aβ deposition and accurately classified Aβ-positive participants (PET: AUC: 94%-97%, cutpoint≥.338 pg/mL; CSF: AUC = .84, cutpoint≥.307 pg/mL). DISCUSSION Plasma p-tau217 is an accurate indicator of amyloid pathology in a heterogeneous cohort, and superior to other plasma biomarkers assessed. Longitudinal analyses assessing impact of comorbidities on p-tau217 utility for disease progression are underway.
Collapse
Affiliation(s)
- Marc D. Rudolph
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Courtney L. Sutphen
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Thomas C. Register
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Samuel N. Lockhart
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Melissa M. Rundle
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Timothy M. Hughes
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - James R. Bateman
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Kiran K. Solingapuram Sai
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Christopher T. Whitlow
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Suzanne Craft
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | - Michelle M. Mielke
- Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| |
Collapse
|
5
|
Ji L, Zhang J. Complex interactions and composite burden of risk factors in vascular cognitive impairment. J Neurol Sci 2025; 468:123367. [PMID: 39733713 DOI: 10.1016/j.jns.2024.123367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/23/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
Vascular cognitive impairment (VCI) stresses the vascular contributions to cognitive decline, ranging from mild to major forms. Except for symptomatic treatment for relevant vascular diseases, the other recommended strategy is to intervene in key vascular risk factors (VRFs) as early as possible. A considerable amount of previous research delineated the association of a specific factor with dementia, involving each risk factor discussed in the present review. However, due to the heterogeneity and complexity of VCI, managing a single factor is insufficient to reduce its incidence and prevalence. Ongoing studies suggest differences in the impact of various combinations of risk factors on dementia. Here in this review, we aimed to provide an updated overview of clinical evidence and implications of complex interactions among various risk factors of VCI, including common VRFs and modifiable dementia-related risk factors. Understating the effect of comorbid risk factors on VCI and underlying mechanisms of them during VCI progression is essential for identifying high-risk population and developing preventive strategies. Furthermore, we summarized common composite risk scores and models used for risk evaluation and prediction of VCI, involving conventional risk scores, subclinical vascular composites, and novel risk models driven by intelligent algorithms. Lastly, we discussed potential gaps and research directions on searching specific clinical risk profiles, constructing effective risk scores, and implementing targeted risk interventions.
Collapse
Affiliation(s)
- Linna Ji
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Huang LY, Ge YJ, Fu Y, Zhao YL, Ou YN, Zhang Y, Ma LZ, Chen SD, Guo ZX, Feng JF, Cheng W, Tan L, Yu JT. Identifying modifiable factors and their joint effect on brain health: an exposome-wide association study. GeroScience 2024; 46:6257-6268. [PMID: 38822946 PMCID: PMC11493923 DOI: 10.1007/s11357-024-01224-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: 12/22/2023] [Accepted: 05/24/2024] [Indexed: 06/03/2024] Open
Abstract
Considerable uncertainty remains regarding the associations of multiple factors with brain health. We aimed to conduct an exposome-wide association study on neurodegenerative disease and neuropsychiatry disorders using data of participants from the UK Biobank. Multivariable Cox regression models with the least absolute shrinkage and selection operator technique as well as principal component analyses were used to evaluate the exposures in relation to common disorders of central nervous system (CNS). Restricted cubic splines were conducted to explore potential nonlinear correlations. Then, weighted standardized scores were generated based on the coefficients to calculate the joint effects of risk factors. We also estimated the potential impact of eliminating the unfavorable profiles of risk domains on CNS disorders using population attributable fraction (PAF). Finally, sensitivity analyses were performed to reduce the risk of reverse causality. The current study discovered the significantly associated exposures fell into six primary exposome categories. The joint effects of identified risk factors demonstrated higher risks for common disorders of CNS (HR = 1.278 ~ 3.743, p < 2e-16). The PAF varied by exposome categories, with lifestyle and medical history contributing to majority of disease cases. In total, we estimated that up to 3.7 ~ 64.1% of disease cases could be prevented.This study yielded modifiable variables of different categories and assessed their joint effects on common disorders of CNS. Targeting the identified exposures might help formulate effective strategies for maintaining brain health.
Collapse
Affiliation(s)
- Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi-Jun Ge
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yong-Li Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling-Zhi Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Xin Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Wei Cheng
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Rudolph MD, Sutphen CL, Register TC, Whitlow CT, Solingapuram Sai KK, Hughes TM, Bateman JR, Dage JL, Russ KA, Mielke MM, Craft S, Lockhart SN. Associations among plasma, MRI, and amyloid PET biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities in a community-dwelling cohort. Alzheimers Dement 2024; 20:4159-4173. [PMID: 38747525 PMCID: PMC11180870 DOI: 10.1002/alz.13835] [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: 11/08/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION We evaluated associations between plasma and neuroimaging-derived biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities. METHODS We examined plasma biomarkers (neurofilament light chain, glial fibrillary acidic protein, amyloid beta [Aβ] 42/40, phosphorylated tau 181) and neuroimaging measures of amyloid deposition (Aβ-positron emission tomography [PET]), total brain volume, white matter hyperintensity volume, diffusion-weighted fractional anisotropy, and neurite orientation dispersion and density imaging free water. Participants were adjudicated as cognitively unimpaired (CU; N = 299), mild cognitive impairment (MCI; N = 192), or dementia (DEM; N = 65). Biomarkers were compared across groups stratified by diagnosis, sex, race, and APOE ε4 carrier status. General linear models examined plasma-imaging associations before and after adjusting for demographics (age, sex, race, education), APOE ε4 status, medications, diagnosis, and other factors (estimated glomerular filtration rate [eGFR], body mass index [BMI]). RESULTS Plasma biomarkers differed across diagnostic groups (DEM > MCI > CU), were altered in Aβ-PET-positive individuals, and were associated with poorer brain health and kidney function. DISCUSSION eGFR and BMI did not substantially impact associations between plasma and neuroimaging biomarkers. HIGHLIGHTS Plasma biomarkers differ across diagnostic groups (DEM > MCI > CU) and are altered in Aβ-PET-positive individuals. Altered plasma biomarker levels are associated with poorer brain health and kidney function. Plasma and neuroimaging biomarker associations are largely independent of comorbidities.
Collapse
Affiliation(s)
- Marc D. Rudolph
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Courtney L. Sutphen
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Thomas C. Register
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher T. Whitlow
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kiran K. Solingapuram Sai
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Timothy M. Hughes
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - James R. Bateman
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jeffrey L. Dage
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kristen A. Russ
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Michelle M. Mielke
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samuel N. Lockhart
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
8
|
Hughes TM, Tanley J, Chen H, Schaich CL, Yeboah J, Espeland MA, Lima JAC, Ambale-Venkatesh B, Michos ED, Ding J, Hayden K, Casanova R, Craft S, Rapp SR, Luchsinger JA, Fitzpatrick AL, Heckbert SR, Post WS, Burke GL. Subclinical vascular composites predict clinical cardiovascular disease, stroke, and dementia: The Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2024; 392:117521. [PMID: 38552474 PMCID: PMC11240239 DOI: 10.1016/j.atherosclerosis.2024.117521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND AND AIMS Subclinical cardiovascular disease (CVD) measures may reflect biological pathways that contribute to increased risk for coronary heart disease (CHD) events, stroke, and dementia beyond conventional risk scores. METHODS The Multi-Ethnic Study of Atherosclerosis (MESA) followed 6814 participants (45-84 years of age) from baseline in 2000-2002 to 2018 over 6 clinical examinations and annual follow-up interviews. MESA baseline subclinical CVD procedures included: seated and supineblood pressure, coronary calcium scan, radial artery tonometry, and carotid ultrasound. Baseline subclinical CVD measures were transformed into z-scores before factor analysis to derive composite factor scores. Time to clinical event for all-cause CVD, CHD, stroke and ICD code-based dementia events were modeled using Cox proportional hazards models reported as area under the curve (AUC) with 95% Confidence Intervals (95%CI) at 10 and 15 years of follow-up. All models included all factor scores together, and adjustment for conventional risk scores for global CVD, stroke, and dementia. RESULTS After factor selection, 24 subclinical measures aggregated into four distinct factors representing: blood pressure, atherosclerosis, arteriosclerosis, and cardiac factors. Each factor significantly predicted time to CVD events and dementia at 10 and 15 years independent of each other and conventional risk scores. Subclinical vascular composites of atherosclerosis and arteriosclerosis best predicted time to clinical events of CVD, CHD, stroke, and dementia. These results were consistent across sex and racial and ethnic groups. CONCLUSIONS Subclinical vascular composites of atherosclerosis and arteriosclerosis may be useful biomarkers to inform the vascular pathways contributing to events of CVD, CHD, stroke, and dementia.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| | - Jordan Tanley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Haiying Chen
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Christopher L Schaich
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Joseph Yeboah
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Mark A Espeland
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Joao A C Lima
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bharath Ambale-Venkatesh
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erin D Michos
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jingzhong Ding
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kathleen Hayden
- Department of Social Sciences and Health Policy, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Ramon Casanova
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Department of Psychiatry and Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Stephen R Rapp
- Department of Psychiatry and Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - José A Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY, United States
| | | | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Wendy S Post
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gregory L Burke
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
9
|
Lachner C, Craver EC, Babulal GM, Lucas JA, Ferman TJ, White RO, Graff-Radford NR, Day GS. Disparate Dementia Risk Factors Are Associated with Cognitive Impairment and Rates of Decline in African Americans. Ann Neurol 2024; 95:518-529. [PMID: 38069571 PMCID: PMC10922775 DOI: 10.1002/ana.26847] [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: 06/03/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVE This study was undertaken to evaluate the frequency of modifiable dementia risk factors and their association with cognitive impairment and rate of decline in diverse participants engaged in studies of memory and aging. METHODS Modifiable dementia risk factors and their associations with cognitive impairment and cognitive decline were determined in community-dwelling African American (AA; n = 261) and non-Hispanic White (nHW; n = 193) participants who completed ≥2 visits at the Mayo Clinic Alzheimer Disease Research Center in Jacksonville, Florida. Risk factors and their associations with cognitive impairment (global Clinical Dementia Rating [CDR] ≥ 0.5) and rates of decline (CDR Sum of Boxes) in impaired participants were compared in AA and nHW participants, controlling for demographics, APOE ɛ4 status, and Area Deprivation Index. RESULTS Hypertension, hypercholesterolemia, obesity, and diabetes were overrepresented in AA participants, but were not associated with cognitive impairment. Depression was associated with increased odds of cognitive impairment in AA (odds ratio [OR] = 4.30, 95% confidence interval [CI] = 2.13-8.67) and nHW participants (OR = 2.79, 95% CI = 1.21-6.44) but uniquely associated with faster decline in AA participants (β = 1.71, 95% CI = 0.69-2.73, p = 0.001). Fewer AA participants reported antidepressant use (9/49, 18%) than nHW counterparts (57/78, 73%, p < 0.001). Vitamin B12 deficiency was also associated with an increased rate of cognitive decline in AA participants (β = 2.65, 95% CI = 0.38-4.91, p = 0.023). INTERPRETATION Modifiable dementia risk factors are common in AA and nHW participants, representing important risk mitigation targets. Depression was associated with dementia in AA and nHW participants, and with accelerated declines in cognitive function in AA participants. Optimizing depression screening and treatment may improve cognitive trajectories and outcomes in AA participants. ANN NEUROL 2024;95:518-529.
Collapse
Affiliation(s)
- Christian Lachner
- Mayo Clinic Florida, Department of Neurology; Jacksonville, FL, 32224, USA
- Mayo Clinic Florida, Department of Psychiatry & Psychology; Jacksonville, FL, 32224, USA
| | - Emily C. Craver
- Mayo Clinic Florida, Department of Quantitative Health Sciences; Jacksonville, FL, 32224, USA
| | - Ganesh M. Babulal
- Washington University in St. Louis, Department of Neurology; St. Louis, MO, 63110, USA
| | - John A. Lucas
- Mayo Clinic Florida, Department of Psychiatry & Psychology; Jacksonville, FL, 32224, USA
| | - Tanis J. Ferman
- Mayo Clinic Florida, Department of Psychiatry & Psychology; Jacksonville, FL, 32224, USA
| | - Richard O. White
- Mayo Clinic Florida, Division of Community Internal Medicine; Jacksonville, FL, 32224, USA
- Mayo Center for Health Equity and Community Engaged Research, Jacksonville, FL, 32224, USA
| | | | - Gregory S. Day
- Mayo Clinic Florida, Department of Neurology; Jacksonville, FL, 32224, USA
| |
Collapse
|
10
|
Zubiagirre U, Ibarrondo O, Larrañaga I, Soto-Gordoa M, Mar-Barrutia L, Mar J. Comorbidity and household income as mediators of gender inequalities in dementia risk: a real-world data population study. BMC Geriatr 2024; 24:209. [PMID: 38424518 PMCID: PMC10905946 DOI: 10.1186/s12877-024-04770-3] [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] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Low household income (HI), comorbidities and female sex are associated with an increased risk of dementia. The aim of this study was to measure the mediating effect of comorbidity and HI on the excess risk due to gender in relation to the incidence and prevalence of dementia in the general population. METHODS A retrospective and observational study using real-world data analysed all people over 60 who were registered with the Basque Health Service in Gipuzkoa. The study measured HI level, the Charlson comorbidity index (CCI), age and sex. The prevalence and incidence of dementia were analysed using logistic regression and Poisson regression models, respectively, adjusted by HI, sex, comorbidity and age. We estimated the combined mediation effect of HI and comorbidity on the prevalence of dementia associated with gender. RESULTS Of the 221,777 individuals, 3.85% (8,549) had a diagnosis of dementia as of 31 December 2021. Classification by the CCI showed a gradient with 2.90% in CCI 0-1, 10.60% in CCI 2-3 and 18.01% in CCI > 3. Both low HI and gender were associated with a higher crude prevalence of dementia. However, in the CCI-adjusted model, women had an increased risk of dementia, while HI was no longer statistically significant. The incidence analysis produced similar results, although HI was not significant in any model. The CCI was significantly higher for men and for people with low HI. The mediation was statistically significant, and the CCI and HI explained 79% of the gender effect. CONCLUSIONS Comorbidity and low HI act as mediators in the increased risk of dementia associated with female sex. Given the difference in the prevalence of comorbidities by HI, individual interventions to control comorbidities could not only prevent dementia but also reduce inequalities, as the risk is greater in the most disadvantaged population.
Collapse
Affiliation(s)
- Uxue Zubiagirre
- Biodonostia Health Research Institute, Donostia-San Sebastián, Guipúzcoa, Spain
- Basque Health Service (Osakidetza), Debagoiena Integrated Healthcare Organisation, Research Unit, Arrasate-Mondragón, Guipúzcoa, Spain
| | - Oliver Ibarrondo
- Biodonostia Health Research Institute, Donostia-San Sebastián, Guipúzcoa, Spain
| | - Igor Larrañaga
- Kronikgune Institute for Health Service Research, Barakaldo, Spain
| | - Myriam Soto-Gordoa
- Faculty of Engineering, Electronics and Computing Department, Mondragon Unibertsitatea, Mondragon, Gipuzkoa, Spain
| | - Lorea Mar-Barrutia
- Department of Psychiatry, Osakidetza Basque Health Service, Araba University Hospital, Vitoria- Gasteiz, Spain
| | - Javier Mar
- Biodonostia Health Research Institute, Donostia-San Sebastián, Guipúzcoa, Spain.
- Basque Health Service (Osakidetza), Debagoiena Integrated Healthcare Organisation, Research Unit, Arrasate-Mondragón, Guipúzcoa, Spain.
- Kronikgune Institute for Health Service Research, Barakaldo, Spain.
| |
Collapse
|
11
|
Myburgh PJ, Solingapuram Sai KK. Two decades of [ 11C]PiB synthesis, 2003-2023: a review. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:48-62. [PMID: 38500746 PMCID: PMC10944378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/04/2024] [Indexed: 03/20/2024]
Abstract
Because carbon-11 (11C) radiotracers cannot be shipped over long distances, their use in routine positron emission tomography (PET) studies is dependent on the production capabilities of individual radiochemistry laboratories. Since 2003, 11C-labeled Pittsburgh compound B ([11C]PiB) has been the gold standard PET radiotracer for in vivo imaging of amyloid β (Aβ) plaques. For more than two decades, researchers have been working to develop faster, higher-yielding, more robust, and optimized production methods with higher radiochemical yields for various imaging applications. This review evaluates progress in [11C]PiB radiochemistry. An introductory overview assesses how it has been applied in clinical neurologic imaging research. We examine the varying approaches reported for radiolabeling, purification, extraction, and formulation. Further considerations for QC methods, regulatory considerations, and optimizations were also discussed.
Collapse
Affiliation(s)
- Paul Josef Myburgh
- Translational Imaging Program, Wake Forest School of MedicineWinston-Salem, NC 27157, USA
| | - Kiran Kumar Solingapuram Sai
- Translational Imaging Program, Wake Forest School of MedicineWinston-Salem, NC 27157, USA
- Department of Radiology, Wake Forest School of MedicineWinston-Salem, NC 27157, USA
| |
Collapse
|
12
|
Ong SS, Peavey JJ, Hiatt KD, Whitlow CT, Sappington RM, Thompson AC, Lockhart SN, Chen H, Craft S, Rapp SR, Fitzpatrick AL, Heckbert SR, Luchsinger JA, Klein BEK, Meuer SM, Cotch MF, Wong TY, Hughes TM. Association of fractal dimension and other retinal vascular network parameters with cognitive performance and neuroimaging biomarkers: The Multi-Ethnic Study of Atherosclerosis (MESA). Alzheimers Dement 2024; 20:941-953. [PMID: 37828734 PMCID: PMC10916935 DOI: 10.1002/alz.13498] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/16/2023] [Accepted: 09/09/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Retinal vascular network changes may reflect the integrity of the cerebral microcirculation, and may be associated with cognitive impairment. METHODS Associations of retinal vascular measures with cognitive function and MRI biomarkers were examined amongst Multi-Ethnic Study of Atherosclerosis (MESA) participants in North Carolina who had gradable retinal photographs at Exams 2 (2002 to 2004, n = 313) and 5 (2010 to 2012, n = 306), and detailed cognitive testing and MRI at Exam 6 (2016 to 2018). RESULTS After adjustment for covariates and multiple comparisons, greater arteriolar fractal dimension (FD) at Exam 2 was associated with less isotropic free water of gray matter regions (β = -0.0005, SE = 0.0024, p = 0.01) at Exam 6, while greater arteriolar FD at Exam 5 was associated with greater gray matter cortical volume (in mm3 , β = 5458, SE = 20.17, p = 0.04) at Exam 6. CONCLUSION Greater arteriolar FD, reflecting greater complexity of the branching pattern of the retinal arteries, is associated with MRI biomarkers indicative of less neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Sally S. Ong
- Department of OphthalmologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jeremy J. Peavey
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kevin D. Hiatt
- Department of RadiologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher T. Whitlow
- Department of RadiologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Rebecca M. Sappington
- Department of OphthalmologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of BiochemistryWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Atalie C. Thompson
- Department of OphthalmologyWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samuel N. Lockhart
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Haiying Chen
- Department of Psychiatry and Behavioral MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Stephen R. Rapp
- Biostatistics and Data ScienceWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Annette L. Fitzpatrick
- Department of EpidemiologySchool of Public HealthUniversity of WashingtonSeattleWashingtonUSA
| | - Susan R. Heckbert
- Department of EpidemiologySchool of Public HealthUniversity of WashingtonSeattleWashingtonUSA
| | - José A. Luchsinger
- Departments of Medicine and EpidemiologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Barbara E. K. Klein
- Department of Ophthalmology and Visual SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Stacy M Meuer
- Department of Ophthalmology and Visual SciencesUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | | | - Tien Y. Wong
- Singapore Eye Research InstituteSingapore National Eye CenterOphthalmology and Visual Sciences Academic Clinical ProgramDuke‐NUS Medical SchoolSingapore
- Tsinghua MedicineTsinghua UniversityBeijingChina
| | - Timothy M. Hughes
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
13
|
Guo ZX, Liu F, Wang FY, Ou YN, Huang LY, Hu H, Wang ZB, Fu Y, Gao PY, Tan L, Yu JT. CAIDE Score, Alzheimer's Disease Pathology, and Cognition in Cognitively Normal Adults: The CABLE Study. J Alzheimers Dis 2024; 99:1273-1283. [PMID: 38728186 DOI: 10.3233/jad-240005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
Background Cardiovascular Risk Factors, Ageing and Dementia (CAIDE) risk score serves as a credible predictor of an individual's risk of dementia. However, studies on the link of the CAIDE score to Alzheimer's disease (AD) pathology are scarce. Objective To explore the links of CAIDE score to cerebrospinal fluid (CSF) biomarkers of AD as well as to cognitive performance. Methods In the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study, we recruited 600 cognitively normal participants. Correlations between the CAIDE score and CSF biomarkers of AD as well as cognitive performance were probed through multiple linear regression models. Whether the correlation between CAIDE score and cognitive performance was mediated by AD pathology was researched by means of mediation analyses. Results Linear regression analyses illustrated that CAIDE score was positively associated with tau-related biomarkers, including pTau (p < 0.001), tTau (p < 0.001), as well as tTau/Aβ42 (p = 0.008), while it was in negative association with cognitive scores, consisting of MMSE score (p < 0.001) as well as MoCA score (p < 0.001). The correlation from CAIDE score to cognitive scores was in part mediated by tau pathology, with a mediation rate varying from 3.2% to 13.2%. Conclusions A higher CAIDE score, as demonstrated in our study, was linked to more severe tau pathology and poorer cognitive performance, and tau pathology mediated the link of CAIDE score to cognitive performance. Increased dementia risk will lead to cognitive decline through aggravating neurodegeneration.
Collapse
Affiliation(s)
- Ze-Xin Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Fang Liu
- Shandong Xiehe University, Jinan, Shandong, China
| | - Fang-Yuan Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Kota KJ, Dawson A, Papas J, Sotelo V, Su G, Li M, Lee W, Estervil J, Marquez M, Sarkar S, Lopez LL, Hu WT. Factors associated with attitudes toward research MRI in older Asian Americans. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12449. [PMID: 38356478 PMCID: PMC10865479 DOI: 10.1002/trc2.12449] [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: 09/13/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 02/16/2024]
Abstract
INTRODUCTION South Asian (SA) and East Asian (EA) older adults represent the fastest-growing racial/ethnic groups of Americans at risk for dementia. While recruiting older SA adults into a brain health study, we encountered unexpected hesitancy toward structural brain magnetic resonance imaging (MRI) analysis and stigmatizing attitudes related to internal locus of control (LoC) for future dementia risks. We hypothesized that support for MRI-related research was influenced by these attitudes as well as personal MRI experience, perceived MRI safety, and concerns for personal risk for future dementia/stroke. METHODS We developed a brief cross-sectional survey to assess older adults' MRI experiences and perceptions, desire to learn of six incidental findings of increasing impact on health, and attitudes related to dementia (including LoC) and research participation. We recruited a convenience sample of 256 respondents (74% reporting as 50+) from the New Jersey/New York City area to complete the survey (offered in English, Chinese, Korean, and Spanish) and modeled the proportional odds (PO) for favorable attitudes toward research activities. RESULTS Seventy-seven SA and 84 EA respondents were analyzed alongside 95 White, Black, or Hispanic adults. White (PO = 2.54, p = 0.013) and EA (PO = 2.14, p = 0.019) respondents were both more likely than SA respondents to endorse healthy volunteers' participation in research, and the difference between White and SA respondents was mediated by the latter's greater internal LoC for dementia risks. EA respondents had more worries for future dementia/stroke than SA respondents (p = 0.006) but still shared SA respondents' lower wish (measured by proportion of total) to learn of incidental MRI findings. DISCUSSION SA-and EA compared to SA-older adults had low desire to learn of incidental MRI findings but had different attitudes toward future dementia/stroke risks. A culturally appropriate protocol to disclose incidental MRI findings may improve SA and EA participation in brain health research. Highlights Older Asian Americans have limited interest in incidental findings on research MRISouth Asians are most likely to attribute dementia to people's own behaviorsSouth Asians' attitudes mediate lower support for healthy volunteers in researchSouth and East Asians differ in dementia worries and research-related attitudes.
Collapse
Affiliation(s)
- Karthik J Kota
- Departments of NeurologyInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
- Departments of MedicineRutgers‐Robert Wood Johnson Medical SchoolInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Alice Dawson
- Departments of NeurologyInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Julia Papas
- Departments of NeurologyInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Victor Sotelo
- Departments of NeurologyInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Guibin Su
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Mei‐Ling Li
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Woowon Lee
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Jaunis Estervil
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Melissa Marquez
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Shromona Sarkar
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - Lisa Lanza Lopez
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| | - William T. Hu
- Departments of NeurologyInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
- Center for Healthy AgingInstitute for Health, Health Care Policy, and Aging ResearchRutgers Biomedical and Health SciencesNew BrunswickNew JerseyUSA
| |
Collapse
|
15
|
Myburgh PJ, Moore MD, Pathirannahel BL, Grace LR, Solingapuram Sai KK. Fully automated production of [ 11C]PiB for clinical use on Trasis-AllinOne synthesizer module. Appl Radiat Isot 2023; 202:111040. [PMID: 37788544 PMCID: PMC10727203 DOI: 10.1016/j.apradiso.2023.111040] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023]
Abstract
Pittsburgh compound B ([11C]PiB) was the first broadly applied radiotracer with specificity for amyloid-β (Aβ) peptide aggregates in the brain and has since been established as the gold standard for positron emission tomography (PET) employed for clinical in vivo imaging of Aβ plaques, used for imaging applications of Alzheimer's disease (AD), related dementia, and other tauopathies. The use of [11C]PiB for routine PET studies is dependent on the production capabilities of each radiochemistry laboratory, subsequently a continuous effort is made to develop suitable and sustainable methods on a variety of auto synthesis platforms. Here we report a fully automated, multi-step radio synthesis, purification, and reformulation of [11C]PiB for PET imaging using the Trasis AllinOne synthesis unit, a commonly used commercial radiochemistry module. We performed three validation runs to evaluate the reproducibility and to verify that the acceptable criteria were met for the release of clinical-grade [11C]PiB using a Trasis AllinOne auto radiosynthesis unit. Solid phase supported radiolabeling was performed through the capture of precursor (6-OH-BTA-0) on a C18 solid phase extraction (SPE) cartridge and subsequent flushing of gaseous [11C]Methyl triflate(MeOTf) through the Sep-Pak for carbon-11 (11C) N-methylation. Starting with 92.5 GBq [11C]CO2, [11C]PiB synthesis was completed in approximately 25 min after cyclotron end of bombardment with an injectable dose >7.0 GBq at end of the synthesis. The radiopharmaceutical product met all quality control criteria and specifications for use in human studies.
Collapse
Affiliation(s)
- Paul Josef Myburgh
- Translational Imaging Program, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Michael David Moore
- Translational Imaging Program, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | | | - Laura Rose Grace
- Translational Imaging Program, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Kiran Kumar Solingapuram Sai
- Translational Imaging Program, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA; Department of Radiology, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
16
|
Zhang Q, Wu G, Zhang X, Wang S, Wang Y. A two-sample Mendelian randomization study of atherosclerosis and dementia. iScience 2023; 26:108325. [PMID: 38026222 PMCID: PMC10654222 DOI: 10.1016/j.isci.2023.108325] [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: 06/27/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The causality between atherosclerosis and dementia remains unclear. This study aimed to explore the causal effect of atherosclerosis related indicators on dementia risk based on two-sample Mendelian randomization (MR) using summary statistics of genome-wide association studies (GWASs). The inverse variance weighted (IVW) method was performed as the main analysis, supplemented by different sensitivity analyses. Suggestive evidence indicated that peripheral arterial disease (PAD) (odds ratio (OR): 0.864, 95% confidence interval (CI): 0.797-0.937), coronary atherosclerosis (CoAS) (OR: 0.927, 95% CI: 0.860-0.998) and atherosclerosis, excluding cerebral, coronary, and PAD (ATHSCLE) (OR: 0.812, 95% CI: 0.725-0.909) were inversely associated with the risk of AD. The sensitivity analysis confirmed a suggestive reverse effect of ATHSCLE on the risk of frontotemporal dementia (FTD) (OR, 0.812, 95% CI, 0.725-0.909). Findings provide suggestive evidence that PAD, CoAS, and ATHSCLE might be associated with the risk of AD or FTD, which requires further exploration in larger samples.
Collapse
Affiliation(s)
- Qiaoyun Zhang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Public Health, Capital Medical University, Beijing, China
| | - Guangheng Wu
- School of Public Health, Capital Medical University, Beijing, China
| | - Xiaoyu Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Youxin Wang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
17
|
Huang LK, Kuan YC, Lin HW, Hu CJ. Clinical trials of new drugs for Alzheimer disease: a 2020-2023 update. J Biomed Sci 2023; 30:83. [PMID: 37784171 PMCID: PMC10544555 DOI: 10.1186/s12929-023-00976-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 10/04/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, presenting a significant unmet medical need worldwide. The pathogenesis of AD involves various pathophysiological events, including the accumulation of amyloid and tau, neuro-inflammation, and neuronal injury. Clinical trials focusing on new drugs for AD were documented in 2020, but subsequent developments have emerged since then. Notably, the US-FDA has approved Aducanumab and Lecanemab, both antibodies targeting amyloid, marking the end of a nearly two-decade period without new AD drugs. In this comprehensive report, we review all trials listed in clinicaltrials.gov, elucidating their underlying mechanisms and study designs. Ongoing clinical trials are investigating numerous promising new drugs for AD. The main trends in these trials involve pathophysiology-based, disease-modifying therapies and the recruitment of participants in earlier stages of the disease. These trends underscore the significance of conducting fundamental research on pathophysiology, prevention, and intervention prior to the occurrence of brain damage caused by AD.
Collapse
Affiliation(s)
- Li-Kai Huang
- PhD Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 291, Zhong Zheng Road, Zhonghe District, New Taipei City, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yi-Chun Kuan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Ho-Wei Lin
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chaur-Jong Hu
- PhD Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, No. 291, Zhong Zheng Road, Zhonghe District, New Taipei City, Taiwan.
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City, Taiwan.
- Dementia Center and Department of Neurology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
18
|
Luo J, Ma Y, Agboola FJ, Grant E, Morris JC, McDade E, Fagan AM, Benzinger TLS, Hassenstab J, Bateman RJ, Perrin RJ, Gordon BA, Goyal M, Strain JF, Yakushev I, Day GS, Xiong C. Longitudinal Relationships of White Matter Hyperintensities and Alzheimer Disease Biomarkers Across the Adult Life Span. Neurology 2023; 101:e164-e177. [PMID: 37202169 PMCID: PMC10351551 DOI: 10.1212/wnl.0000000000207378] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 03/20/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND AND OBJECTIVES White matter hyperintensities (WMH) correlate with Alzheimer disease (AD) biomarkers cross-sectionally and modulate AD pathogenesis. Longitudinal changes have been reported for AD biomarkers, including concentrations of CSF β-amyloid (Aβ) 42, Aβ40, total tau and phosphorylated tau181, standardized uptake value ratio from the molecular imaging of cerebral fibrillar Aβ with PET using [11C] Pittsburgh Compound-B, MRI-based hippocampal volume, and cortical thickness. Correlations between established AD biomarkers and the longitudinal change for WMH have not been fully evaluated, especially among cognitively normal individuals across the adult life span. METHODS We jointly analyzed the longitudinal data of WMH volume and each of the established AD biomarkers and cognition from 371 cognitively normal individuals whose baseline age spanned from 19.6 to 88.20 years from 4 longitudinal studies of aging and AD. A 2-stage algorithm was applied to identify the inflection point of baseline age whereby older participants had an accelerated longitudinal change in WMH volume, in comparison with the younger participants. The longitudinal correlations between WMH volume and AD biomarkers were estimated from the bivariate linear mixed-effects models. RESULTS A longitudinal increase in WMH volume was associated with a longitudinal increase in PET amyloid uptake and a decrease in MRI hippocampal volume, cortical thickness, and cognition. The inflection point of baseline age in WMH volume was identified at 60.46 (95% CI 56.43-64.49) years, with the annual increase for the older participants (83.12 [SE = 10.19] mm3 per year) more than 13 times faster (p < 0.0001) than that for the younger participants (6.35 [SE = 5.63] mm3 per year). Accelerated rates of change among the older participants were similarly observed in almost all the AD biomarkers. Longitudinal correlations of WMH volume with MRI, PET amyloid biomarkers, and cognition seemed to be numerically stronger for the younger participants, but not significantly different from those for the older participants. Carrying APOE ε4 alleles did not alter the longitudinal correlations between WMH and AD biomarkers. DISCUSSION Longitudinal increases in WMH volume started to accelerate around a baseline age of 60.46 years and correlated with the longitudinal change in PET amyloid uptake, MRI structural outcomes, and cognition.
Collapse
Affiliation(s)
- Jingqin Luo
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Yinjiao Ma
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Folasade Jane Agboola
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Elizabeth Grant
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - John C Morris
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Eric McDade
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Anne M Fagan
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Tammie L S Benzinger
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Jason Hassenstab
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Randall J Bateman
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Richard J Perrin
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Brian A Gordon
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Manu Goyal
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Jeremy F Strain
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Igor Yakushev
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Gregory S Day
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL
| | - Chengjie Xiong
- From the Division of Public Health Sciences (J.L.), Department of Surgery, Siteman Cancer Center Biostatistics Core (J.L.), Division of Biostatistics (J.L., Y.M., F.J.A., E.G., C.X.), Knight Alzheimer Disease Research Center (Y.M., F.J.A., E.G., J.C.M., A.M.F., T.L.S.B., J.H., R.J.B., R.J.P., B.A.G., C.X.), Department of Neurology (J.C.M., E.M., A.M.F., J.H., R.J.B., R.J.P., M.G., J.F.S.), Department of Pathology and Immunology (J.C.M., R.J.P.), and Department of Radiology (T.L.S.B., B.A.G., M.G.), Washington University School of Medicine, St. Louis, MO; Department of Nuclear Medicine (I.Y.), and Klinikum rechts der Isar (I.Y.), School of Medicine, Neuroimaging Center (TUM-NIC), Technical University of Munich, Germany; and Department of Neurology (G.S.D.), Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
19
|
Elahi FM, Alladi S, Black SE, Claassen JAHR, DeCarli C, Hughes TM, Moonen J, Pajewski NM, Price BR, Satizabal C, Shaaban CE, Silva NCBS, Snyder HM, Sveikata L, Williamson JD, Wolters FJ, Hainsworth AH. Clinical trials in vascular cognitive impairment following SPRINT-MIND: An international perspective. Cell Rep Med 2023; 4:101089. [PMID: 37343515 PMCID: PMC10314118 DOI: 10.1016/j.xcrm.2023.101089] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/19/2022] [Accepted: 05/24/2023] [Indexed: 06/23/2023]
Abstract
A large interventional trial, the Systolic Blood Pressure Intervention Trial sub-study termed Memory and Cognition in Decreased Hypertension (SPRINT-MIND), found reduced risk of cognitive impairment in older adults with intensive, relative to standard, blood-pressure-lowering targets (systolic BP < 120 vs. <140 mm Hg). In this perspective, we discuss key questions and make recommendations for clinical practice and for clinical trials, following SPRINT-MIND. Future trials should embody cognitive endpoints appropriate to the participant group, ideally with adaptive designs that ensure robust answers for cognitive and cardiovascular endpoints. Reliable data from diverse populations, including the oldest-old (age > 80 years), will maximize external validity and global implementation of trial findings. New biomarkers will improve phenotyping to stratify patients to optimal treatments. Currently no antihypertensive drug class stands out for dementia risk reduction. Multi-domain interventions, incorporating lifestyle change (exercise, diet) alongside medications, may maximize global impact. Given the low cost and wide availability of antihypertensive drugs, intensive BP reduction may be a cost-effective means to reduce dementia risk in diverse, aging populations worldwide.
Collapse
Affiliation(s)
- Fanny M Elahi
- Friedman Brain Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Suvarna Alladi
- National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka 560030, India
| | - Sandra E Black
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Jurgen A H R Claassen
- Department of Geriatric Medicine and Donders Institute for Medical Neuroscience, Radboud University Medical Center, 6525 EN Nijmegen, the Netherlands
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA 95817, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Justine Moonen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, the Netherlands
| | - Nicholas M Pajewski
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC 27154, USA
| | | | - Claudia Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX 78229, USA
| | - C Elizabeth Shaaban
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nárlon C B S Silva
- Djavad Mowafaghian Centre for Brain Health, Department of Physical Therapy, Faculty of Medicine, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Heather M Snyder
- Alzheimer's Association, 225 N Michigan Avenue, Chicago, IL 60603, USA
| | - Lukas Sveikata
- J.P. Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, 1205 Genève, Switzerland; Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jeff D Williamson
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27154, USA
| | - Frank J Wolters
- Departments of Epidemiology and Radiology & Nuclear Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 GD Rotterdam, the Netherlands
| | - Atticus H Hainsworth
- Neurology, St George's University Hospitals NHS Foundation Trust, London SW17 0QT, UK; Molecular and Clinical Sciences Research Institute, St George's University of London, London SW17 0RE, UK.
| |
Collapse
|
20
|
Hughes TM, Tanley J, Chen H, Schaich CL, Yeboah J, Espeland MA, Lima JAC, Ambale-Venkatesh B, Michos ED, Ding J, Hayden K, Casanova R, Craft S, Rapp SR, Luchsinger JA, Fitzpatrick AL, Heckbert SR, Post WS, Burke GL. Subclinical Vascular Composites Predict Clinical Cardiovascular Disease, Stroke, and Dementia: The Multi-Ethnic Study of Atherosclerosis (MESA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.01.23289364. [PMID: 37205504 PMCID: PMC10187443 DOI: 10.1101/2023.05.01.23289364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Background Subclinical cardiovascular disease (CVD) measures may reflect biological pathways that contribute to increased risk for coronary heart disease (CHD) events, stroke, and dementia beyond conventional risk scores. Methods The Multi-Ethnic Study of Atherosclerosis (MESA) followed 6,814 participants (45-84 years of age) from baseline in 2000-2002 to 2018 over 6 clinical examinations and annual follow-up interviews. MESA baseline subclinical CVD procedures included: seated and supine blood pressure, coronary calcium scan, radial artery tonometry, and carotid ultrasound. Baseline subclinical CVD measures were transformed into z-scores before factor analysis to derive composite factor scores. Time to clinical event for all CVD, CHD, stroke and ICD code-based dementia events were modeled using Cox proportional hazards models reported as area under the curve (AUC) with 95% Confidence Intervals (95%CI) at 10 and 15 years of follow-up. All models included all factor scores together and adjustment for conventional risk scores for global CVD, stroke, and dementia. Results After factor selection, 24 subclinical measures aggregated into four distinct factors representing: blood pressure, arteriosclerosis, atherosclerosis, and cardiac factors. Each factor significantly predicted time to CVD events and dementia at 10 and 15 years independent of each other and conventional risk scores. Subclinical vascular composites of arteriosclerosis and atherosclerosis best predicted time to clinical events of CVD, CHD, stroke, and dementia. These results were consistent across sex and racial and ethnic groups. Conclusions Subclinical vascular composites of arteriosclerosis and atherosclerosis may be useful biomarkers to inform the vascular pathways contributing to events of CVD, CHD, stroke, and dementia.
Collapse
|
21
|
Kota K, Dawson A, Papas J, Sotelo V, Su G, Li ML, Lee W, Estervil J, Marquez M, Sarkar S, Lopez LL, Hu WT. Too much information? Asian Americans' preferences for incidental brain MRI findings. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.17.23288629. [PMID: 37162874 PMCID: PMC10168418 DOI: 10.1101/2023.04.17.23288629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
INTRODUCTION South Asian (SA) and East Asian (EA) older adults represent the fastest growing group of Americans at risk for dementia, but their participation in aging and dementia research has been limited. While recruiting healthy SA older adults into a brain health study, we encountered unexpected hesitancy towards structural brain MRI analysis along with some stigmatizing attitudes related to internal locus of control (LoC) for future dementia risks. We hypothesized that support for MRI-related research was influenced by these attitudes as well as one's own MRI experience, perceived MRI safety, and concerns for one's own risks for future dementia/stroke. METHODS We developed a brief cross-sectional survey to assess older adults' MRI experiences and perceptions, desire to learn of six incidental findings of increasing health implications, and attitudes related to dementia as well as research participation. We recruited a convenience sample of 256 respondents (74% reporting as 50+) from the New Jersey/New York City area to complete the survey, and modeled the proportional odds (P.O.) for pro-research attitudes. RESULTS 77 SA and 84 EA respondents were analyzed with 95 non-Asian adults. White (P.O.=2.54, p=0.013) and EA (P.O.=2.14, p=0.019) respondents were both more likely than SA respondents to endorse healthy volunteers' participation in research, and the difference between White and SA respondents was mediated by the latter's greater internal LoC for dementia risks. EA respondents had more worries for future dementia/stroke than SA respondents (p=0.006), but still shared SA respondents' low desire to learn of incidental MRI findings. DISCUSSION SA and EA older adults had different attitudes towards future dementia/stroke risks, but shared a low desire to learn of incidental MRI findings. A culturally-appropriate protocol to disclose incidental MRI findings may improve SA and EA participation in brain health research.
Collapse
Affiliation(s)
- Karthik Kota
- Department of Neurology Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Alice Dawson
- Department of Neurology Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Julia Papas
- Department of Neurology Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Victor Sotelo
- Department of Neurology Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Guibin Su
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Mei-Ling Li
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Woowon Lee
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Jaunis Estervil
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Melissa Marquez
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Shromona Sarkar
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - Lisa Lanza Lopez
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| | - William T. Hu
- Department of Neurology Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
- Department of Center for Healthy Aging, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, 08901
| |
Collapse
|
22
|
Wang Q, Xu S, Liu F, Liu Y, Liu Y, Xu F. Development of risk prediction model for cognitive impairment in patients with coronary heart disease: A study protocol for a prospective, cross-sectional analysis. Front Cardiovasc Med 2023; 9:1107544. [PMID: 36727026 PMCID: PMC9885763 DOI: 10.3389/fcvm.2022.1107544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
Background Ischemic heart disease and degenerative encephalopathy are two main sources of disease burden for the global elderly population. Coronary heart disease (CHD) and cognitive impairment, as representative diseases, are prevalent and serious illnesses in the elderly. According to recent research, patients with CHD are more likely to experience cognitive impairment and their cognitive ability declines more quickly. Vascular risk factors have been associated with differences in cognitive performance in epidemiological studies, but evidence in patients with CHD is more limited. Inextricably linked between the heart and the brain. Considering the unique characteristics of recurrent cognitive impairment in patients with CHD, we will further study the related risk factors. We tried to investigate the potential predictors of cognitive impairment in patients with CHD through a prospective, cross-sectional study. Methods The cross-sectional study design will recruit 378 patients with CHD (≥65 years) from Xiyuan Hospital of China Academy of Chinese Medical Sciences. The subjects' cognitive function is evaluated with MoCA scale, and they are divided into cognitive impairment group and normal cognitive function group according to the score results. Demographic data, disease characteristics (results of coronary CT/ angiography, number of stents implanted, status of diseased vessels), laboratory tests (biochemistry, coagulation, serum iron levels, pulse wave velocity), metabolites (blood samples and intestinal metabolites), and lifestyle (smoking, alcohol consumption, sleep, physical activity) will be assessed as outcome indicators. Compare the two groups and the correlation analysis will be performed on the development of mild cognitive impairment. Mann-Whitney U or X2 test was selected to describe and evaluate the variation, and logistics regression analysis was employed to fit the prediction model. After that, do the calibration curve and decision curve to evaluate the model. The prediction model will be validated by a validation set. Discussion To explore the risk factors related to mild cognitive impairment (MCI) in patients with CHD, a new predictive model is established, which can achieve advanced intervention in the occurrence of MCI after CHD. Owing to its cross-sectional study design, the study has some limitations, but it will be further studied by increasing the observation period, adding follow-up data collection or prospective cohort study. The study has been registered with the China Clinical Trials Registry (ChiCTR2200063255) to conduct clinical trials.
Collapse
Affiliation(s)
- Qing Wang
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Xu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fenglan Liu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfei Liu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Yue Liu ✉
| | - Fengqin Xu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Fengqin Xu ✉
| |
Collapse
|
23
|
Akushevich I, Kravchenko J, Yashkin A, Doraiswamy PM, Hill CV. Expanding the scope of health disparities research in Alzheimer's disease and related dementias: Recommendations from the "Leveraging Existing Data and Analytic Methods for Health Disparities Research Related to Aging and Alzheimer's Disease and Related Dementias" Workshop Series. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12415. [PMID: 36935764 PMCID: PMC10020680 DOI: 10.1002/dad2.12415] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
Topics discussed at the "Leveraging Existing Data and Analytic Methods for Health Disparities Research Related to Aging and Alzheimer's Disease and Related Dementias" workshop, held by Duke University and the Alzheimer's Association with support from the National Institute on Aging, are summarized. Ways in which existing data resources paired with innovative applications of both novel and well-known methodologies can be used to identify the effects of multi-level societal, community, and individual determinants of race/ethnicity, sex, and geography-related health disparities in Alzheimer's disease and related dementia are proposed. Current literature on the population analyses of these health disparities is summarized with a focus on identifying existing gaps in knowledge, and ways to mitigate these gaps using data/method combinations are discussed at the workshop. Substantive and methodological directions of future research capable of advancing health disparities research related to aging are formulated.
Collapse
Affiliation(s)
- Igor Akushevich
- Social Science Research InstituteBiodemography of Aging Research UnitDuke UniversityDurhamNorth CarolinaUSA
| | - Julia Kravchenko
- Duke University School of MedicineDepartment of SurgeryDurhamNorth CarolinaUSA
| | - Arseniy Yashkin
- Social Science Research InstituteBiodemography of Aging Research UnitDuke UniversityDurhamNorth CarolinaUSA
| | - P. Murali Doraiswamy
- Departments of Psychiatry and MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | | | | |
Collapse
|
24
|
Bouges S, Fischer B, Norton DL, Wyman MF, Lambrou N, Zuelsdorff M, Van Hulle CA, Ennis GE, James TT, Johnson AL, Chin N, Carlsson CM, Gleason CE. Effect of Metabolic Syndrome Risk Factors on Processing Speed and Executive Function in Three Racialized Groups. J Alzheimers Dis 2023; 92:285-294. [PMID: 36744341 PMCID: PMC10211459 DOI: 10.3233/jad-220920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Metabolic syndrome (MetS) has been associated with increased risk for Alzheimer's disease and related dementias (ADRD). Understanding the association of MetS risk factors to processing speed and executive function in the pre-clinical stages of ADRD in under-represented groups would offer insight on potential mechanisms through which MetS associates with ADRD risk. OBJECTIVE Examine association of MetS features and processing speed and executive function across three racial groups. METHODS Cognitively unimpaired adults from the Wisconsin Alzheimer's Disease Research Center and the Wisconsin Registry for Alzheimer's Disease Prevention completed blood-draws and neuropsychological testing. Six cognitive outcomes were assessed in association to MetS risk factors: Trailmaking Tests A and B, Animal Fluency, Digit Symbol, and composite scores for Processing Speed and Executive Function. Linear mixed effect models were used to assess the relationship between MetS risk factor count and longitudinal cognitive performance across three racialized groups. RESULTS Participant sample sizes varied by outcome analyzed (N = 714-1,088). African American and Native American groups exhibited higher rates of MetS than non-Hispanic Whites. MetS was associated with processing speed and executive function across all racialized groups. Three-way interaction by racialized group was limited to one cognitive outcome: Trailmaking Test A. CONCLUSION Metabolic dysfunction incrementally affects cognitive trajectory, with generally similar associations across racial groups. Since racialized groups exhibit higher levels of both MetS and ADRD, MetS may represent a driving factor for increased ADRD risk experience by racialized group and an important and modifiable target through which to reduce risk of ADRD.
Collapse
Affiliation(s)
- Shenikqua Bouges
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
| | - Barbara Fischer
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Division of Neurology, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Derek L. Norton
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Mary F. Wyman
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
- University of Wisconsin School of Medicine & Public Health, Department of Psychiatry, Madison, WI, USA
| | - Nickolas Lambrou
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
| | - Megan Zuelsdorff
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, UW School of Medicine & Public Health, Madison, WI, USA
- University of Wisconsin – Madison School of Nursing
| | - Carol A. Van Hulle
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
| | - Gilda E. Ennis
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
| | - Taryn T. James
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
| | - Adrienne L. Johnson
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- University of Wisconsin School of Medicine & Public Health, Center for Tobacco Research and Intervention, Madison, WI, USA
| | - Nathaniel Chin
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, UW School of Medicine & Public Health, Madison, WI, USA
| | - Cynthia M. Carlsson
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, UW School of Medicine & Public Health, Madison, WI, USA
| | - Carey E. Gleason
- VA Geriatric Research, Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin (UW) School of Medicine & Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, UW School of Medicine & Public Health, Madison, WI, USA
| |
Collapse
|
25
|
Abstract
This review deals with an unwelcome reality about several forms of dementia, including Alzheimer's disease- that these dementias are caused, in part or whole, by the aging of the vasculature. Since the vasculature ages in us all, dementia is our fate, sealed by the realit!ies of the circulation; it is not a disease with a cure pending. Empirically, cognitive impairment before our 7th decade is uncommon and considered early, while a diagnosis in our 11th decade is late but common in that cohort (>40%). Projections from earlier ages suggest that the prevalence of dementia in people surviving into their 12th decade exceeds 80%. We address the question why so few of many interventions known to delay dementia are recognized as therapy; and we try to resolve this few-and-many paradox, identifying opportunities for better treatment, especially pre-diagnosis. The idea of dementia as a fate is resisted, we argue, because it negates the hope of a cure. But the price of that hope is lost opportunity. An approach more in line with the evidence, and more likely to limit suffering, is to understand the damage that accumulates with age in the cerebral vasculature and therefore in the brain, and which eventually gives rise to cognitive symptoms in late life, too often leading to dementia. We argue that hope should be redirected to delaying that damage and with it the onset of cognitive loss; and, for each individual, it should be redirected to a life-long defense of their brain.
Collapse
Affiliation(s)
- Marcus J Andersson
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Jonathan Stone
- School of Medical Sciences and Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
26
|
Glymphatic Dysfunction Mediates the Influence of White Matter Hyperintensities on Episodic Memory in Cerebral Small Vessel Disease. Brain Sci 2022; 12:brainsci12121611. [PMID: 36552071 PMCID: PMC9775074 DOI: 10.3390/brainsci12121611] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022] Open
Abstract
Glymphatic dysfunction has been linked to cognitive decline in several neurodegenerative diseases. In cerebral small vessel disease (CSVD), the mechanism of white matter hyperintensities (WMH)-related cognitive impairment (CI) is still under investigation. The diffusion tensor image (DTI) analysis along the perivascular space (ALPS) method has been considered to be a reliable parameter to evaluate glymphatic function. Therefore, we applied the ALPS-index to determine the influence of glymphatic function on CI in CSVD. In total, 137 CSVD patients (normal cognitive group, mild CI group, and dementia group) and 52 normal controls were included in this study. The ALPS-index was calculated based on the DTI. Correlation analyses and mediation analysis were conducted to examine the relationship between glymphatic function and cognition. Remarkable differences in the ALPS-index were observed between subjects with and without CI. The ALPS-index was negatively correlated with age, WMH volume, and general cognitive function in all CSVD patients. In the mild CI group, the ALPS-index was independently positively related to episodic memory, and mediated the relationship between WMH volume and episodic memory. In conclusion, the ALPS-index is a potential marker for early recognition of CI in CSVD. Glymphatic dysfunction mediates the relationship between WMH and CI in CSVD.
Collapse
|
27
|
Kim SE, Kim HJ, Jang H, Weiner MW, DeCarli C, Na DL, Seo SW. Interaction between Alzheimer's Disease and Cerebral Small Vessel Disease: A Review Focused on Neuroimaging Markers. Int J Mol Sci 2022; 23:10490. [PMID: 36142419 PMCID: PMC9499680 DOI: 10.3390/ijms231810490] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of β-amyloid (Aβ) and tau, and subcortical vascular cognitive impairment (SVCI) is characterized by cerebral small vessel disease (CSVD). They are the most common causes of cognitive impairment in the elderly population. Concurrent CSVD burden is more commonly observed in AD-type dementia than in other neurodegenerative diseases. Recent developments in Aβ and tau positron emission tomography (PET) have enabled the investigation of the relationship between AD biomarkers and CSVD in vivo. In this review, we focus on the interaction between AD and CSVD markers and the clinical effects of these two markers based on molecular imaging studies. First, we cover the frequency of AD imaging markers, including Aβ and tau, in patients with SVCI. Second, we discuss the relationship between AD and CSVD markers and the potential distinct pathobiology of AD markers in SVCI compared to AD-type dementia. Next, we discuss the clinical effects of AD and CSVD markers in SVCI, and hemorrhagic markers in cerebral amyloid angiopathy. Finally, this review provides both the current challenges and future perspectives for SVCI.
Collapse
Affiliation(s)
- Si Eun Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan 48108, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Michael W. Weiner
- Center for Imaging of Neurodegenerative Diseases, University of California, San Francisco, CA 94121, USA
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California, Davis, CA 95616, USA
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
28
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|