1
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Liang J, Pan Y, Zhang W, Gao D, Ma J, Zhang Y, Ji M, Dai Y, Liu Y, Wang Y, Zhu Y, Lu B, Xie W, Zheng F. Associations Between Atherosclerosis and Subsequent Cognitive Decline: A Prospective Cohort Study. J Am Heart Assoc 2024; 13:e036696. [PMID: 39494555 DOI: 10.1161/jaha.124.036696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND This study aimed to examine whether baseline atherosclerosis was associated with subsequent short-term domain-specific cognitive decline. METHODS AND RESULTS This research was based on the BRAVE (Beijing Research on Aging and Vessel) study, a population-based prospective cohort study of adults aged 40 to 80 years, free of dementia. At baseline (wave 1, 2019), cognitive assessments and atherosclerosis measures, including carotid intima-media thickness, carotid plaques, coronary artery calcification, and brachial-ankle pulse wave velocity were conducted. Cognitive function was reassessed in wave 2 (2022-2023) using linear mixed models for analysis. A total of 932 participants (63.7% women; mean age, 60.0±6.9 years) were included. Compared with the lowest tertile of carotid intima-media thickness, carotid plaques, and brachial-ankle pulse wave velocity, or a coronary artery calcification score=0, the highest tertile of carotid intima-media thickness (β=-0.065 SD/y [95% CI, -0.112 to -0.017]; P=0.008), carotid plaques (β=-0.070 SD/y [95% CI, -0.130 to -0.011]; P=0.021), and brachial-ankle pulse wave velocity (β=-0.057 SD/y [95% CI, -0.105 to -0.010]; P=0.018), and a coronary artery calcification score≥400 (β=-0.081 SD/y [95% CI, -0.153 to -0.008]; P=0.029) were significantly associated with a faster decline in semantic fluency after multivariable adjustment. Moreover, greater carotid intima-media thickness, coronary artery calcification, and brachial-ankle pulse wave velocity were significantly associated with a faster decline in global cognition. CONCLUSIONS More significant atherosclerosis was associated with faster semantic fluency and global cognition declines.
Collapse
Affiliation(s)
- Jie Liang
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Yang Pan
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Wenya Zhang
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Darui Gao
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Jingya Ma
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Yanyu Zhang
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Mengmeng Ji
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Yiwen Dai
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Yuling Liu
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Yongqian Wang
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Yidan Zhu
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Bin Lu
- Department of Radiology Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- State Key Lab and National Center for Cardiovascular Diseases Beijing China
| | - Wuxiang Xie
- Peking University First Hospital Beijing China
- Clinical Research Institute, Institute of Advanced Clinical Medicine, Peking University Beijing China
- Key Laboratory of Epidemiology of Major Diseases Peking University, Ministry of Education Beijing China
| | - Fanfan Zheng
- School of Nursing Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
3
|
Pan X, Liu Y, Zhou F, Tao Y, Liu R, Tian B, Li N, Chen S, Xing Y. Associations between carotid plaques and white matter hyperintensities in cerebral small vessel disease. J Clin Neurosci 2024; 129:110871. [PMID: 39433006 DOI: 10.1016/j.jocn.2024.110871] [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: 07/15/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) can lead to stroke and cognitive impairment. Small vessels cannot be visualized by neuroimaging directly, and CSVD can only be evaluated by cerebral parenchymal changes in MRI. Therefore, a convenient screening method for identifying high-risk and susceptible patients is needed. Recently, some studies found that CSVD was related to large atherosclerosis, and atherosclerosis was an essential pathological feature of CSVD. Therefore, we aimed to investigate the association between carotid plaque size characteristics and white matter hyperintensities (WMHs) in patients with CSVD. METHODS We continuously enrolled patients with CSVD. Carotid plaque features were evaluated using carotid ultrasound, and WMHs were evaluated using brain magnetic resonance imaging. Plaque characteristics were compared between patients with no/mild WMHs and those with severe WMHs. Associations between the plaque characteristics and WMH severity were analyzed using logistic regression. RESULTS In total, 180 patients were recruited, of whom 92 had severe WMHs. The severe WMHs group had a higher sum of the bilateral maximum intima-media thickness (4.15 mm vs. 3.30 mm), longer maximum plaque length (17.20 mm vs. 13.90 mm), thicker plaques (2.70 mm vs. 2.30 mm), and more plaques (3 vs. 2) than the no/mild WMHs group. Adjusted logistic regression analyses revealed that maximum plaque length and thickness were associated with WMHs. CONCLUSIONS Carotid atherosclerotic plaque features, such as plaque length and thickness, were associated with the severity of WMHs, which suggested that carotid atherosclerotic plaque characteristics measured using ultrasound might be helpful indicators for identifying high-risk patients with CSVD.
Collapse
Affiliation(s)
- Xijuan Pan
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yumei Liu
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Fubo Zhou
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yunlu Tao
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Ran Liu
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Bing Tian
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Na Li
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Songwei Chen
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yingqi Xing
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China; Beijing Diagnostic Center of Vascular Ultrasound, Beijing, China; Center of Vascular Ultrasonography, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Deng T, Yu W, Lü Y. Different physical exercise in the treatment of Alzheimer's disease. Psychogeriatrics 2024. [PMID: 39460576 DOI: 10.1111/psyg.13207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Alzheimer's disease (AD) is rapidly becoming one of the most expensive, burdening, and deadly diseases of this century. Up to now, there is still a lack of pharmacotherapy with substantial efficacy, and physical exercise is a promising and low-cost way to aid in delaying the process of AD. The aim of this review is to summarise the efficacy of different physical exercise approaches and doses in the management of AD, including aerobic exercise, resistance exercise and multicomponent exercise. A literature search using MeSH terms for each topic is undertaken using PubMed and the Web of Science Core Collection database, supplemented by hand searching for additional references. Retrieved articles were reviewed, synthesised, and summarised. This review shows that aerobic exercise has been almost unanimously recognised for the improvement of cognition, neuropsychiatric symptoms and activities of daily living. Resistance exercise also shows a good prospect in the above aspects and has a unique advantage in enhancing muscle strength, while multicomponent physical activity does not seen to significantly improve cognitive function. Each type of exercise has a dose effect, but for individuals with AD, the most appropriate exercise dose is still controversial.
Collapse
Affiliation(s)
- Tianqing Deng
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weihua Yu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Csiszar A, Ungvari A, Patai R, Gulej R, Yabluchanskiy A, Benyo Z, Kovacs I, Sotonyi P, Kirkpartrick AC, Prodan CI, Liotta EM, Zhang XA, Toth P, Tarantini S, Sorond FA, Ungvari Z. Atherosclerotic burden and cerebral small vessel disease: exploring the link through microvascular aging and cerebral microhemorrhages. GeroScience 2024; 46:5103-5132. [PMID: 38639833 PMCID: PMC11336042 DOI: 10.1007/s11357-024-01139-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Cerebral microhemorrhages (CMHs, also known as cerebral microbleeds) are a critical but frequently underestimated aspect of cerebral small vessel disease (CSVD), bearing substantial clinical consequences. Detectable through sensitive neuroimaging techniques, CMHs reveal an extensive pathological landscape. They are prevalent in the aging population, with multiple CMHs often being observed in a given individual. CMHs are closely associated with accelerated cognitive decline and are increasingly recognized as key contributors to the pathogenesis of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD). This review paper delves into the hypothesis that atherosclerosis, a prevalent age-related large vessel disease, extends its pathological influence into the cerebral microcirculation, thereby contributing to the development and progression of CSVD, with a specific focus on CMHs. We explore the concept of vascular aging as a continuum, bridging macrovascular pathologies like atherosclerosis with microvascular abnormalities characteristic of CSVD. We posit that the same risk factors precipitating accelerated aging in large vessels (i.e., atherogenesis), primarily through oxidative stress and inflammatory pathways, similarly instigate accelerated microvascular aging. Accelerated microvascular aging leads to increased microvascular fragility, which in turn predisposes to the formation of CMHs. The presence of hypertension and amyloid pathology further intensifies this process. We comprehensively overview the current body of evidence supporting this interconnected vascular hypothesis. Our review includes an examination of epidemiological data, which provides insights into the prevalence and impact of CMHs in the context of atherosclerosis and CSVD. Furthermore, we explore the shared mechanisms between large vessel aging, atherogenesis, microvascular aging, and CSVD, particularly focusing on how these intertwined processes contribute to the genesis of CMHs. By highlighting the role of vascular aging in the pathophysiology of CMHs, this review seeks to enhance the understanding of CSVD and its links to systemic vascular disorders. Our aim is to provide insights that could inform future therapeutic approaches and research directions in the realm of neurovascular health.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Illes Kovacs
- Department of Ophthalmology, Semmelweis University, 1085, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Angelia C Kirkpartrick
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xin A Zhang
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
An W, Zhou J, Qiu Z, Wang P, Han X, Cheng Y, He Z, An Y, Li S. Identification of crosstalk genes and immune characteristics between Alzheimer's disease and atherosclerosis. Front Immunol 2024; 15:1443464. [PMID: 39188714 PMCID: PMC11345154 DOI: 10.3389/fimmu.2024.1443464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Background Advancements in modern medicine have extended human lifespan, but they have also led to an increase in age-related diseases such as Alzheimer's disease (AD) and atherosclerosis (AS). Growing research evidence indicates a close connection between these two conditions. Methods We downloaded four gene expression datasets related to AD and AS from the Gene Expression Omnibus (GEO) database (GSE33000, GSE100927, GSE44770, and GSE43292) and performed differential gene expression (DEGs) analysis using the R package "limma". Through Weighted gene correlation network analysis (WGCNA), we selected the gene modules most relevant to the diseases and intersected them with the DEGs to identify crosstalk genes (CGs) between AD and AS. Subsequently, we conducted functional enrichment analysis of the CGs using DAVID. To screen for potential diagnostic genes, we applied the least absolute shrinkage and selection operator (LASSO) regression and constructed a logistic regression model for disease prediction. We established a protein-protein interaction (PPI) network using STRING (https://cn.string-db.org/) and Cytoscape and analyzed immune cell infiltration using the CIBERSORT algorithm. Additionally, NetworkAnalyst (http://www.networkanalyst.ca) was utilized for gene regulation and interaction analysis, and consensus clustering was employed to determine disease subtypes. All statistical analyses and visualizations were performed using various R packages, with a significance level set at p<0.05. Results Through intersection analysis of disease-associated gene modules identified by DEGs and WGCNA, we identified a total of 31 CGs co-existing between AD and AS, with their biological functions primarily associated with immune pathways. LASSO analysis helped us identify three genes (C1QA, MT1M, and RAMP1) as optimal diagnostic CGs for AD and AS. Based on this, we constructed predictive models for both diseases, whose accuracy was validated by external databases. By establishing a PPI network and employing four topological algorithms, we identified four hub genes (C1QB, CSF1R, TYROBP, and FCER1G) within the CGs, closely related to immune cell infiltration. NetworkAnalyst further revealed the regulatory networks of these hub genes. Finally, defining C1 and C2 subtypes for AD and AS respectively based on the expression profiles of CGs, we found the C2 subtype exhibited immune overactivation. Conclusion This study utilized gene expression matrices and various algorithms to explore the potential links between AD and AS. The identification of CGs revealed interactions between these two diseases, with immune and inflammatory imbalances playing crucial roles in their onset and progression. We hope these findings will provide valuable insights for future research on AD and AS.
Collapse
Affiliation(s)
- Wenhao An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jiajun Zhou
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Zhiqiang Qiu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Peishen Wang
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Xinye Han
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yanwen Cheng
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Zi He
- Department of Research and Development, Beijing Yihua Biotechnology Co., Ltd, Beijing, China
| | - Yihua An
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Ceccon M, Kantsjö JB, Ronchi F. Personalized Paths: Unlocking Alzheimer's via the Gut-Brain Axis. Visc Med 2024; 40:194-209. [PMID: 39157730 PMCID: PMC11326767 DOI: 10.1159/000535869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/14/2023] [Indexed: 08/20/2024] Open
Abstract
Background Alzheimer's disease (AD) is characterised by abnormal protein aggregates in the brain that lead to cognitive decline. While current therapies only treat symptoms, disease-modifying treatments are urgently needed. Studies suggest that the composition of the microbiota is altered in people with AD, suggesting a link between gut bacteria and AD-related brain changes. Summary In our narrative review, we explore various microbial interventions, such as faecal microbiota transplantation, probiotics, and diet, as powerful potential treatments. Studies suggest changes in microbiota composition following these interventions, with some beneficial effects on cognitive function. However, the mechanism of action of these microbial interventions is still unknown. Key Message Our aim was to highlight the importance of personalised approaches, taking into account individual metabolic and microbiome profiles. We try to address gaps in current research and emphasise the need for microbiota analysis at different stages of the disease and its integration with clinical parameters and lifestyle information for a comprehensive understanding of AD progression (summarised in online suppl. Fig. 1; for all online suppl. material, see https://doi.org/10.1159/000535869).
Collapse
Affiliation(s)
- Matteo Ceccon
- Institute of Microbiology, Infectious Disease, and Immunology/Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johan B Kantsjö
- Institute of Microbiology, Infectious Disease, and Immunology/Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Francesca Ronchi
- Institute of Microbiology, Infectious Disease, and Immunology/Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
8
|
Chen YH, Ren CY, Yu C. Causal relationship between Alzheimer's disease and unstable angina: a bidirectional Mendelian randomization analysis. Front Psychiatry 2024; 15:1435394. [PMID: 39045549 PMCID: PMC11263098 DOI: 10.3389/fpsyt.2024.1435394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Background Research from observational studies has demonstrated a link between Alzheimer's disease (AD) and a higher risk of cardiovascular disease (CVD). Uncertainty surrounds the exact genetic cause of AD and coronary heart disease, particularly unstable angina (UA). Mendelian randomization (MR) analysis was used to examine the causal genetic link between AD and UA to evaluate the impact of AD on UA. Methods The purpose of the bidirectional MR analysis was to investigate the link between exposure and illness causation. Genetic instrumental variables for AD were obtained from European populations using genome-wide association studies (GWAS). The primary causal conclusions were obtained using the inverse variance weighted approach (IVW), and other sensitivity analysis techniques were employed. Sensitivity analyses were carried out to evaluate heterogeneity and horizontal pleiotropy to guarantee accurate MR results. Results An elevated risk of UA was linked to genetically predicted AD (IVW: OR=3.439, 95% CI: 1.565-7.555, P=0.002). A substantial genetic relationship between UA and the risk of AD was not supported by any evidence in the reverse study (IVW: OR=0.998, 95% CI: 0.995-1.001, P=0.190). Various MR techniques produced consistent results. Sensitivity analysis revealed no discernible heterogeneity or horizontal pleiotropy. Conclusions One risk factor for UA that we found in our bidirectional Mendelian randomization trial was AD. This highlights the necessity of researching the underlying molecular mechanisms linked to AD and UA as well as the possibility of creating individualized treatment plans based on genetic data.
Collapse
Affiliation(s)
- Yu-hang Chen
- Department of Operations Management, Chongqing Mental Health Center, Chongqing, China
| | - Cong-ying Ren
- Department of Hospital Infection Control, Chongqing Mental Health Center, Chongqing, China
| | - Cao Yu
- Department of Cardiothoracic Surgery, Chongqing University Jiangjin Hospital, Chongqing, China
| |
Collapse
|
9
|
Wójcik P, Jastrzębski MK, Zięba A, Matosiuk D, Kaczor AA. Caspases in Alzheimer's Disease: Mechanism of Activation, Role, and Potential Treatment. Mol Neurobiol 2024; 61:4834-4853. [PMID: 38135855 PMCID: PMC11236938 DOI: 10.1007/s12035-023-03847-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
With the aging of the population, treatment of conditions emerging in old age, such as neurodegenerative disorders, has become a major medical challenge. Of these, Alzheimer's disease, leading to cognitive dysfunction, is of particular interest. Neuronal loss plays an important role in the pathophysiology of this condition, and over the years, a great effort has been made to determine the role of various factors in this process. Unfortunately, until now, the exact pathomechanism of this condition remains unknown. However, the most popular theories associate AD with abnormalities in the Tau and β-amyloid (Aβ) proteins, which lead to their deposition and result in neuronal death. Neurons, like all cells, die in a variety of ways, among which pyroptosis, apoptosis, and necroptosis are associated with the activation of various caspases. It is worth mentioning that Tau and Aβ proteins are considered to be one of the caspase activators, leading to cell death. Moreover, the protease activity of caspases influences both of the previously mentioned proteins, Tau and Aβ, converting them into more toxic derivatives. Due to the variety of ways caspases impact the development of AD, drugs targeting caspases could potentially be useful in the treatment of this condition. Therefore, there is a constant need to search for novel caspase inhibitors and evaluate them in preclinical and clinical trials.
Collapse
Affiliation(s)
- Piotr Wójcik
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland.
| | - Michał K Jastrzębski
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Agata Zięba
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Dariusz Matosiuk
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Agnieszka A Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland.
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
10
|
Morgan AE, Mc Auley MT. Vascular dementia: From pathobiology to emerging perspectives. Ageing Res Rev 2024; 96:102278. [PMID: 38513772 DOI: 10.1016/j.arr.2024.102278] [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: 01/26/2024] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Vascular dementia (VaD) is the second most common type of dementia. VaD is synonymous with ageing, and its symptoms place a significant burden on the health and wellbeing of older people. Despite the identification of a substantial number of risk factors for VaD, the pathological mechanisms underpinning this disease remain to be fully elucidated. Consequently, a biogerontological imperative exists to highlight the modifiable lifestyle factors which can mitigate against the risk of developing VaD. This review will critically examine some of the factors which have been revealed to modulate VaD risk. The survey commences by providing an overview of the putative mechanisms which are associated with the pathobiology of VaD. Next, the factors which influence the risk of developing VaD are examined. Finally, emerging treatment avenues including epigenetics, the gut microbiome, and pro-longevity pharmaceuticals are discussed. By drawing this key evidence together, it is our hope that it can be used to inform future experimental investigations in this field.
Collapse
Affiliation(s)
- Amy Elizabeth Morgan
- School of Health and Sports Sciences, Hope Park, Liverpool Hope University, Liverpool L16 9JD, United Kingdom.
| | - Mark Tomás Mc Auley
- School of Science, Engineering and Environment, University of Salford Manchester, Salford M5 4NT, United Kingdom
| |
Collapse
|
11
|
Li M, Yin Y, Qin D. Treadmill training impacts the skeletal muscle molecular clock after ischemia stroke in rats. Heliyon 2024; 10:e27430. [PMID: 38509905 PMCID: PMC10951531 DOI: 10.1016/j.heliyon.2024.e27430] [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: 05/14/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Objective Stroke is frequently associated with muscle mass loss. Treadmill training is considered the most effective treatment for sarcopenia. Circadian rhythms are closely related to exercise and have been extensively studied. The skeletal muscle has its molecular clock genes. Exercise may regulate skeletal muscle clock genes. This study evaluated the effects of early treadmill training on the skeletal muscle molecular clock machinery in rats with stroke and determined the relationship of these changes with exercise-induced improvements in skeletal muscle health. Materials and methods Overall, 168 Sprague-Dawley rats were included in this study. We established an ischemic stroke rat model of sarcopenia. Finally, 144 rats were randomly allocated to four groups (36 per group): normal, sham, middle cerebral artery occlusion, and training. Neurological scores, rotating rod test, body weight, muscle circumference, wet weight, and hematoxylin-eosin staining were assessed. Twenty-four rats were used for transcriptome sequencing. Gene and protein expressions of skeletal muscles, such as brain muscle arnt-like 1, period 1, and period 2, were measured by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assays. Results Neurological function scores and rotating rod test results improved after treadmill training. Nine differentially expressed genes were identified by comparing the sham group with the hemiplegic side of the model group. Seventeen differentially expressed genes were identified between the hemiplegic and non-hemiplegic sides. BMAL1, PER1, and PER2 mRNA levels increased on both sides after treadmill training. BMAL1 expression increased, and PER1 expression decreased on both sides, whereas PER2 expression decreased on the hemiplegic side but increased on the non-hemiplegic side. Conclusion Treadmill training can mitigate muscle loss and regulate skeletal muscle clock gene expression following ischemic stroke. Exercise affects the hemiplegic side and has a positive regulatory effect on the non-hemiplegic side.
Collapse
Affiliation(s)
- Mai Li
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, No. 374, Fengning Street, Dianmian Road, 650101, Kunming, China
| | - Yong Yin
- Department of Rehabilitation Medicine, The Affiliated Hospital of Yunnan University, No. 176, Qingnian Road, 650021, Kunming, China
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, No. 1076 Yuhua Road, Chenggong District, 650500, Kunming, China
| |
Collapse
|
12
|
Luo J, Thomassen JQ, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. Neutrophil counts and cardiovascular disease. Eur Heart J 2023; 44:4953-4964. [PMID: 37950632 PMCID: PMC10719495 DOI: 10.1093/eurheartj/ehad649] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND AND AIMS Anti-inflammatory trials have shown considerable benefits for cardiovascular disease. High neutrophil counts, an easily accessible inflammation biomarker, are associated with atherosclerosis in experimental studies. This study aimed to investigate the associations between neutrophil counts and risk of nine cardiovascular endpoints using observational and genetic approaches. METHODS Observational studies were conducted in the Copenhagen General Population Study (n = 101 730). Genetic studies were firstly performed using one-sample Mendelian randomization (MR) with individual-level data from the UK Biobank (n = 365 913); secondly, two-sample MR analyses were performed using summary-level data from the Blood Cell Consortium (n = 563 085). Outcomes included ischaemic heart disease, myocardial infarction, peripheral arterial disease, ischaemic cerebrovascular disease, ischaemic stroke, vascular-related dementia, vascular dementia, heart failure, and atrial fibrillation. RESULTS Observational analyses showed associations between high neutrophil counts with high risks of all outcomes. In the UK Biobank, odds ratios (95% confidence intervals) per 1-SD higher genetically predicted neutrophil counts were 1.15 (1.08, 1.21) for ischaemic heart disease, 1.22 (1.12, 1.34) for myocardial infarction, and 1.19 (1.04, 1.36) for peripheral arterial disease; similar results were observed in men and women separately. In two-sample MR, corresponding estimates were 1.14 (1.05, 1.23) for ischaemic heart disease and 1.11 (1.02, 1.20) for myocardial infarction; multiple sensitivity analyses showed consistent results. No robust associations in two-sample MR analyses were found for other types of leucocytes. CONCLUSIONS Observational and genetically determined high neutrophil counts were associated with atherosclerotic cardiovascular disease, supporting that high blood neutrophil counts is a causal risk factor for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Jiao Luo
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Jesper Qvist Thomassen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital-Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Sun W, Zhuo S, Wu H, Cai X. Association between Coronary Heart Disease, Heart Failure, and Risk of Alzheimer's Disease: A Systematic Review and Meta-Analysis. Ann Indian Acad Neurol 2023; 26:958-965. [PMID: 38229651 PMCID: PMC10789403 DOI: 10.4103/aian.aian_361_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/02/2023] [Accepted: 06/24/2023] [Indexed: 01/18/2024] Open
Abstract
Background Cardiovascular diseases such as coronary heart disease (CHD), heart failure (HF), and stroke have been linked to the development of Alzheimer's disease (AD). However, previous studies have reported inconsistent results. The study aimed to investigate the association between CHD, HF, and the risk of AD using a meta-analysis. Methods STATA 12.0 software is used to compute odds ratios (ORs)/relative risks (RRs) and 95% confidence intervals (CIs) for the association between CHD, HF, and the risk of AD. Results A total of 12 studies (including N = 36,913 individuals with AD and N = 1,701,718 participants) investigated the association between CHD and the risk of AD. Meta-analysis indicated that CHD was associated with an increased risk of AD with a random effects model (OR/RR: 1.22, 95% CI: 1.00-1.48, I2 = 97.2%, P < 0.001). Additionally, seven studies (including N = 5,119 individuals with AD and N = 1,231,399 participants) investigated the association between myocardial infarction (MI) and the risk of AD. Our meta-analysis demonstrated no significant association between MI and the risk of AD with a fixed effects model (RR: 1.09, 95% CI: 0.91-1.30, I2 = 42.8%, P = 0.105). Finally, six studies (including N = 83,065 individuals with AD and N = 2,414,963 participants) examined the association between HF and the risk of AD. Our meta-analysis revealed that HF was associated with an increased risk of AD using a random effects model (RR: 1.53, 95% CI: 1.05-2.24, I2 = 96.8%, P < 0.001). Conclusion In conclusion, our meta-analysis suggests that CHD and HF are associated with an increased risk of developing AD. Nonetheless, more large-scale prospective studies are necessary to further investigate the associations between CHD, HF, and the risk of AD.
Collapse
Affiliation(s)
- Weifeng Sun
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Shanshan Zhuo
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Hao Wu
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xiaojie Cai
- Department of Cardiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
14
|
Hoost SS, Brickman AM, Manly JJ, Honig LS, Gu Y, Sanchez D, Reyes-Dumeyer D, Lantigua RA, Kang MS, Dage JL, Mayeux R. Effects of Vascular Risk Factors on the Association of Blood-Based Biomarkers with Alzheimer's Disease. MEDICAL RESEARCH ARCHIVES 2023; 11:10.18103/mra.v11i9.4468. [PMID: 38037598 PMCID: PMC10688358 DOI: 10.18103/mra.v11i9.4468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Background Comorbidities may influence the levels of blood-based biomarkers for Alzheimer's disease (AD). We investigated whether differences in risk factors or comorbid conditions might explain the discordance between clinical diagnosis and biomarker classifications in a multi-ethnic cohort of elderly individuals. Aims To evaluate the relationship of medical conditions and other characteristics, including body mass index (BMI), vascular risk factors, and head injury, with cognitive impairment and blood-based biomarkers of AD, phosphorylated tau (P-tau 181, P-tau 217), in a multi-ethnic cohort. Methods Three-hundred individuals, aged 65 and older, were selected from a prospective community-based cohort for equal representation among three racial/ethnic groups: non-Hispanic White, Hispanic/Latino and African American/Black. Participants were classified into four groups based on absence (Asym) or presence (Sym) of cognitive impairment and low (NEG) or high (POS) P-tau 217 or P-tau 181 levels, determined previously in the same cohort: (Asym/NEG, Asym/POS, Sym/NEG, Sym/POS). We examined differences in individual characteristics across the four groups. We performed post-hoc analysis examining the differences across biomarker and cognitive status. Results P-tau 217 or P-tau 181 positive individuals had lower BMI than P-tau negative participants, regardless of symptom status. Symptomatic and asymptomatic participants did not differ in terms of BMI. BMI was not a mediator of the effect of P-tau 217 or P-tau 181 on dementia. Frequencies of other risk factors did not differ between the four groups of individuals. Conclusions Participants with higher levels of P-tau 217 or P-tau 181 consistent with AD had lower BMI regardless of whether the individual was symptomatic. These findings suggest that weight loss may change with AD biomarker levels before onset of cognitive decline. They do not support BMI as a confounding variable. Further longitudinal studies could explore the relationship of risk factors with clinical diagnoses and biomarkers.
Collapse
Affiliation(s)
- SS Hoost
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - AM Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - JJ Manly
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - LS Honig
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - Y Gu
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - D Sanchez
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - D Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - RA Lantigua
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| | - MS Kang
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - JL Dage
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine Indianapolis IN
| | - R Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, New York, NY
| |
Collapse
|
15
|
Llido JP, Jayanti S, Tiribelli C, Gazzin S. Bilirubin and Redox Stress in Age-Related Brain Diseases. Antioxidants (Basel) 2023; 12:1525. [PMID: 37627520 PMCID: PMC10451892 DOI: 10.3390/antiox12081525] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Cellular redox status has a crucial role in brain physiology, as well as in pathologic conditions. Physiologic senescence, by dysregulating cellular redox homeostasis and decreasing antioxidant defenses, enhances the central nervous system's susceptibility to diseases. The reduction of free radical accumulation through lifestyle changes, and the supplementation of antioxidants as a prophylactic and therapeutic approach to increase brain health, are strongly suggested. Bilirubin is a powerful endogenous antioxidant, with more and more recognized roles as a biomarker of disease resistance, a predictor of all-cause mortality, and a molecule that may promote health in adults. The alteration of the expression and activity of the enzymes involved in bilirubin production, as well as an altered blood bilirubin level, are often reported in neurologic conditions and neurodegenerative diseases (together denoted NCDs) in aging. These changes may predict or contribute both positively and negatively to the diseases. Understanding the role of bilirubin in the onset and progression of NCDs will be functional to consider the benefits vs. the drawbacks and to hypothesize the best strategies for its manipulation for therapeutic purposes.
Collapse
Affiliation(s)
- John Paul Llido
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
- Department of Science and Technology, Philippine Council for Health Research and Development, Bicutan, Taguig City 1631, Philippines
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
| | - Sri Jayanti
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16911, Indonesia
| | - Claudio Tiribelli
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
| | - Silvia Gazzin
- Liver Brain Unit “Rita Moretti”, Italian Liver Foundation, Bldg. Q, AREA Science Park, Basovizza, 34149 Trieste, Italy; (J.P.L.); or (S.J.); (S.G.)
| |
Collapse
|
16
|
Thurston RC, Wu M, Barinas-Mitchell E, Chang Y, Aizenstein H, Derby CA, Maki PM. Carotid intima media thickness and white matter hyperintensity volume among midlife women. Alzheimers Dement 2023; 19:3129-3137. [PMID: 36722746 PMCID: PMC10390649 DOI: 10.1002/alz.12951] [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: 08/24/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Carotid atherosclerosis may be associated with brain white matter hyperintensities (WMH). Few studies consider women at midlife, a critical time for women's cardiovascular and brain health. We tested the hypothesis that higher carotid intima media thickness (IMT) would be associated with greater WMH volume (WMHV) among midlife women. We explored interactions by apolipoprotein E (APOE) ε4 status. METHODS Two hundred thirty-nine women aged 45 to 67 underwent carotid artery ultrasound, phlebotomy, and magnetic resonance imaging (MRI). One hundred seventy participants had undergone an ultrasound 5 years earlier. RESULTS Higher IMT was associated with greater whole brain (B[standard error (SE)] = 0.77 [.31], P = 0.01; multivariable) and periventricular (B[SE] = 0.80 [.30], P = 0.008; multivariable) WMHV. Associations were observed for IMT assessed contemporaneously with the MRI and 5 years prior to the MRI. Associations were strongest for APOE ε4-positive women. DISCUSSION Among midlife women, higher IMT was associated with greater WMHV. Vascular risk is critical to midlife brain health, particularly for APOE ε4-positive women.
Collapse
Affiliation(s)
- Rebecca C. Thurston
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA USA
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | | | - Yuefang Chang
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA USA
| | - Howard Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Carol A. Derby
- Department of Neurology, and Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pauline M. Maki
- Departments of Psychiatry, Psychology, and Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, IL USA
| |
Collapse
|
17
|
Xia H, Jin B, Su C, Zhao K, Ma R. Early diagnostic value of intima-media thickness and D-dimer levels for vascular dementia. Medicine (Baltimore) 2023; 102:e34149. [PMID: 37352042 PMCID: PMC10289637 DOI: 10.1097/md.0000000000034149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/07/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023] Open
Abstract
To evaluate the diagnostic value of combination of D-dimer and Doppler Intima-Media Thickness (IMT) based on clinical data for vascular dementia (VaD). In this study, 100 cases of initial VaD were diagnosed by Zhenjiang Fourth People Hospital as VaD group, and 100 healthy were engaged as control group, medical history and test results were collected for evaluation. IMT and D-dimer were highly correlated with the onset of VaD. The sensitivity of combination diagnosis for early VaD: 90.2%, the specificity: 87.9%, the area under the curve (AUC) is 0.872, which were superior to IMT or D-dimer alone. D-dimer and IMT are significantly increased in the initial onset of cerebrovascular diseases, and combined detection of them is conducive to early diagnosis and evaluation of the disease. Given the limitations of D-dimer and IMT, combined detection is more conducive to early diagnosis and prognosis, and can be used as screening and routine examination items, even evaluate the severity of cognitive decline.
Collapse
Affiliation(s)
- Haiping Xia
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Biao Jin
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Cen Su
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Kangren Zhao
- Zhenjiang 4th People’s Hospital Affiliated to Jiangsu University, Jiangsu, China
| | - Rui Ma
- Department of Anatomy, Medical School, Jiangsu University, Jiangsu, China
| |
Collapse
|
18
|
Dybjer E, Kumar A, Nägga K, Engström G, Mattsson-Carlgren N, Nilsson PM, Melander O, Hansson O. Polygenic risk of type 2 diabetes is associated with incident vascular dementia: a prospective cohort study. Brain Commun 2023; 5:fcad054. [PMID: 37091584 PMCID: PMC10118265 DOI: 10.1093/braincomms/fcad054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/28/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Type 2 diabetes and dementia are associated, but it is unclear whether the two diseases have common genetic risk markers that could partly explain their association. It is also unclear whether the association between the two diseases is of a causal nature. Furthermore, few studies on diabetes and dementia have validated dementia end-points with high diagnostic precision. We tested associations between polygenic risk scores for type 2 diabetes, fasting glucose, fasting insulin and haemoglobin A1c as exposure variables and dementia as outcome variables in 29 139 adults (mean age 55) followed for 20-23 years. Dementia diagnoses were validated by physicians through data from medical records, neuroimaging and biomarkers in cerebrospinal fluid. The dementia end-points included all-cause dementia, mixed dementia, Alzheimer's disease and vascular dementia. We also tested causal associations between type 2 diabetes and dementia through two-sample Mendelian randomization analyses. Seven different polygenic risk scores including single-nucleotide polymorphisms with different significance thresholds for type 2 diabetes were tested. A polygenic risk score including 4891 single-nucleotide polymorphisms with a P-value of <5e-04 showed the strongest association with different outcomes, including all-cause dementia (hazard ratio 1.11; Bonferroni corrected P = 3.6e-03), mixed dementia (hazard ratio 1.18; Bonferroni corrected P = 3.3e-04) and vascular dementia cases (hazard ratio 1.28; Bonferroni corrected P = 9.6e-05). The associations were stronger for non-carriers of the Alzheimer's disease risk gene APOE ε4. There was, however, no significant association between polygenic risk scores for type 2 diabetes and Alzheimer's disease. Furthermore, two-sample Mendelian randomization analyses could not confirm a causal link between genetic risk markers of type 2 diabetes and dementia outcomes. In conclusion, polygenic risk of type 2 diabetes is associated with an increased risk of dementia, in particular vascular dementia. The findings imply that certain people with type 2 diabetes may, due to their genetic background, be more prone to develop diabetes-associated dementia. This knowledge could in the future lead to targeted preventive strategies in clinical practice.
Collapse
Affiliation(s)
- Elin Dybjer
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
| | - Atul Kumar
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
| | - Katarina Nägga
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
- Department of Acute Internal Medicine and Geriatrics, Linköping University, SE-58183 Linköping, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
- Brain Injury After Cardiac Arrest Research Group, Lund University, Box 117, SE-22100 Lund, Sweden
- WCMM – Wallenberg Centre for Molecular Medicine, Lund University, Sölvegatan 19, BMC D11, SE-22184 Lund, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- EpiHealth: Epidemiology for Health Strategic Research Area, Lund University, SUS Malmö, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- EpiHealth: Epidemiology for Health Strategic Research Area, Lund University, SUS Malmö, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, SE-20502 Malmö, Sweden
- EXODIAB: Excellence in Diabetes Research in Sweden, Lund University, Box 117, SE-22100 Lund, Sweden
| | - Oskar Hansson
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
| |
Collapse
|
19
|
Hendriks S, Peetoom K, Bakker C, Koopmans R, van der Flier W, Papma J, Verhey F, de Vugt M, Köhler S. Global incidence of young-onset dementia: A systematic review and meta-analysis. Alzheimers Dement 2023; 19:831-843. [PMID: 35715891 DOI: 10.1002/alz.12695] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/19/2022] [Accepted: 04/27/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Reliable data on the incidence rates for young-onset dementia (YOD) are lacking, but are necessary for research on disease etiology and to raise awareness among health care professionals. METHODS We performed a systematic review and meta-analysis on population-based studies on the incidence of YOD, published between January 1, 1990 and February 1, 2022, according to Meta-analyses of Observational Studies in Epidemiology (MOOSE) guidelines. Data were analyzed using random-effects meta-analyses. Results were age-standardized, and heterogeneity was assessed by subgroup analyses and meta-regression. RESULTS Sixty-one articles were included. Global age-standardized incidence rates increased from 0.17/100,000 in age 30 to 34 years, to 5.14/100,000 in age 60 to 64 years, giving a global total age-standardized incidence rate of 11 per 100,000 in age 30 to 64. This corresponds to 370,000 new YOD cases annually worldwide. Heterogeneity was high and meta-regression showed geographic location significantly influenced this heterogeneity. DISCUSSION This meta-analysis shows the current best estimate of YOD incidence. New prospective cohort studies are needed.
Collapse
Affiliation(s)
- Stevie Hendriks
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Kirsten Peetoom
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Christian Bakker
- Department of Primary and Community Care, Radboud UMC Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
- Groenhuysen, Center for Specialized Geriatric Care, Roosendaal, The Netherlands
| | - Raymond Koopmans
- Department of Primary and Community Care, Radboud UMC Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wiesje van der Flier
- Department of Neurology, Department of Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Janne Papma
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Marjolein de Vugt
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
20
|
Sergi D, Zauli E, Tisato V, Secchiero P, Zauli G, Cervellati C. Lipids at the Nexus between Cerebrovascular Disease and Vascular Dementia: The Impact of HDL-Cholesterol and Ceramides. Int J Mol Sci 2023; 24:ijms24054403. [PMID: 36901834 PMCID: PMC10002119 DOI: 10.3390/ijms24054403] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cerebrovascular diseases and the subsequent brain hypoperfusion are at the basis of vascular dementia. Dyslipidemia, marked by an increase in circulating levels of triglycerides and LDL-cholesterol and a parallel decrease in HDL-cholesterol, in turn, is pivotal in promoting atherosclerosis which represents a common feature of cardiovascular and cerebrovascular diseases. In this regard, HDL-cholesterol has traditionally been considered as being protective from a cardiovascular and a cerebrovascular prospective. However, emerging evidence suggests that their quality and functionality play a more prominent role than their circulating levels in shaping cardiovascular health and possibly cognitive function. Furthermore, the quality of lipids embedded in circulating lipoproteins represents another key discriminant in modulating cardiovascular disease, with ceramides being proposed as a novel risk factor for atherosclerosis. This review highlights the role of HDL lipoprotein and ceramides in cerebrovascular diseases and the repercussion on vascular dementia. Additionally, the manuscript provides an up-to-date picture of the impact of saturated and omega-3 fatty acids on HDL circulating levels, functionality and ceramide metabolism.
Collapse
Affiliation(s)
- Domenico Sergi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
- Correspondence:
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Veronica Tisato
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- King Khaled Eye Specialistic Hospital, Riyadh 11462, Saudi Arabia
| | - Carlo Cervellati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
21
|
Cerebral amyloid-β deposition in patients with heart disease or carotid occlusive disease: A systematic review and meta-analysis. J Neurol Sci 2023; 445:120551. [PMID: 36669349 DOI: 10.1016/j.jns.2023.120551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Cardiovascular disease is an important contributor to cognitive impairment. This likely involves prototypical vascular disease mechanisms like ischemia, but cardiovascular disease might also impact the brain by accelerating cerebral amyloid-β accumulation. We aimed to determine whether there is an association between heart disease or carotid occlusive disease (COD) and cerebral amyloid-β burden. METHODS We conducted a systematic review of studies investigating cerebral amyloid-β burden, measured with positron emission tomography, in adults with and without heart disease or COD. Where possible, we obtained standardized mean differences (SMD) of amyloid-β standardized uptake volume ratios (SUVr) for meta-analysis. RESULTS Eight cross-sectional studies were identified (1478 participants, aged 60-81 years, 51% female). Three studies on heart disease (two on atrial fibrillation (AF) only, one on AF, coronary artery disease and heart failure) did not find a difference in amyloid-β burden between patients and controls. The pooled difference for 746 participants with and without AF did not reach significance (SMD SUVr 0.14, 95%CI -0.06-0.34). Of the five studies on COD (one on differences between participants with and without COD, four on differences between hemispheres in unilateral COD), four did not find a difference in amyloid-β between participants or hemispheres. The pooled difference in amyloid-β load between hemispheres in 24 patients with unilateral COD was not significant (SMD SUVr -0.13, 95%CI -0.70-0.43). CONCLUSION Based on current studies, although limited and heterogeneous, there is insufficient evidence to support the hypothesis that heart disease or COD are associated with increased cerebral amyloid-β burden.
Collapse
|
22
|
Grant WB, Blake SM. Diet's Role in Modifying Risk of Alzheimer's Disease: History and Present Understanding. J Alzheimers Dis 2023; 96:1353-1382. [PMID: 37955087 PMCID: PMC10741367 DOI: 10.3233/jad-230418] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/14/2023]
Abstract
Diet is an important nonpharmacological risk-modifying factor for Alzheimer's disease (AD). The approaches used here to assess diet's role in the risk of AD include multi-country ecological studies, prospective and cross-sectional observational studies, and laboratory studies. Ecological studies have identified fat, meat, and obesity from high-energy diets as important risk factors for AD and reported that AD rates peak about 15-20 years after national dietary changes. Observational studies have compared the Western dietary pattern with those of the Dietary Approaches to Stop Hypertension (DASH), Mediterranean (MedDi), and Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diets. Those studies identified AD risk factors including higher consumption of saturated and total fats, meat, and ultraprocessed foods and a lower risk of AD with higher consumption of fruits, legumes, nuts, omega-3 fatty acids, vegetables, and whole grains. Diet-induced factors associated with a significant risk of AD include inflammation, insulin resistance, oxidative stress, elevated homocysteine, dietary advanced glycation end products, and trimethylamine N-oxide. The molecular mechanisms by which dietary bioactive components and specific foods affect risk of AD are discussed. Given most countries' entrenched food supply systems, the upward trends of AD rates would be hard to reverse. However, for people willing and able, a low-animal product diet with plenty of anti-inflammatory, low-glycemic load foods may be helpful.
Collapse
Affiliation(s)
- William B. Grant
- Sunlight, Nutrition, and Health Research Center, San Francisco, CA, USA
| | - Steven M. Blake
- Nutritional Neuroscience, Maui Memory Clinic, Wailuku, HI, USA
| |
Collapse
|
23
|
Gu N, Fan Y, Zhou L, Zhang Y, Bai W, Li Y, Yang L, Li J. Influence of Tuina plus oxiracetam on serum inflammatory factors and oxidative stress in mild vascular dementia patients. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2022. [DOI: 10.1007/s11726-022-1324-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
24
|
Dong C, Zhou C, Fu C, Hao W, Ozaki A, Shrestha N, Virani SS, Mishra SR, Zhu D. Sex differences in the association between cardiovascular diseases and dementia subtypes: a prospective analysis of 464,616 UK Biobank participants. Biol Sex Differ 2022; 13:21. [PMID: 35526028 PMCID: PMC9080133 DOI: 10.1186/s13293-022-00431-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/23/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Whether the association of cardiovascular diseases (CVDs) with dementia differs by sex remains unclear, and the role of socioeconomic, lifestyle, genetic, and medical factors in their association is unknown. METHODS We used data from the UK Biobank, a population-based cohort study of 502,649 individuals. We used Cox proportional hazards models to estimate sex-specific hazard ratios (HRs) and 95% confidence intervals (CI), and women-to-men ratio of HRs (RHR) for the association between CVD (coronary heart diseases (CHD), stroke, and heart failure) and incident dementia (all-cause dementia, Alzheimer's Disease (AD), and vascular dementia (VD)). The moderator roles of socioeconomic (education, income), lifestyle (smoking, BMI, leisure activities, and physical activity), genetic factors (APOE allele status), and medical history were also analyzed. RESULTS Compared to people who did not experience a CVD event, the HRs (95%CI) between CVD and all-cause dementia were higher in women compared to men, with an RHR (Female/Male) of 1.20 (1.13, 1.28). Specifically, the HRs for AD were higher in women with CHD and heart failure compared to men, with an RHR (95%CI) of 1.63 (1.39, 1.91) and 1.32 (1.07, 1.62) respectively. The HRs for VD were higher in men with heart failure than women, with RHR (95%CI) of 0.73 (0.57, 0.93). An interaction effect was observed between socioeconomic, lifestyle, genetic factors, and medical history in the sex-specific association between CVD and dementia. CONCLUSION Women with CVD were 1.5 times more likely to experience AD than men, while had 15% lower risk of having VD than men.
Collapse
Affiliation(s)
- Caiyun Dong
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.,NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Chunmiao Zhou
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.,NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Chunying Fu
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.,NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Wenting Hao
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.,NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Fukushima, Japan.,Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Nipun Shrestha
- Department of Primary Care and Mental Health, University of Liverpool, Liverpool, UK
| | - Salim S Virani
- Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, TX, USA
| | - Shiva Raj Mishra
- Academy for Data Sciences and Global Health, Kathmandu, Nepal.,Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Dongshan Zhu
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China. .,NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China. .,Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
25
|
Wang X, Qiao T, Liu M, Wang X. Homocysteine Associated With Low Cognitive Function Independent of Asymptomatic Intracranial and Carotid Arteries Stenoses in Chinese Elderly Patients: An Outpatient-Based Cross-Sectional Study. J Geriatr Psychiatry Neurol 2022; 35:302-308. [PMID: 33504251 DOI: 10.1177/0891988720988914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION A high homocysteine (Hcy) concentration is correlated with cognitive impairment; however, the exact underlying mechanism is still not fully elucidated. The present study aimed to investigate whether asymptomatic intracranial and carotid arteries stenoses are involved in Hcy-related low cognitive function. METHODS This was a cross-sectional study in outpatient clinics. Residents aged ≥60 years, who came to the Stroke and Rehabilitation Clinic of Shandong Provincial Third Hospital in Jinan, Shandong Province from December 2019 to May 2020 to seek consultation due to abnormal transcranial Doppler reports (eg., increased cerebral blood flow velocity) were eligible. Information including demographics, medical history, lifestyle habits were collected. Fasting blood was used to detect total serum homocysteine level (tHcy). Cerebrovascular magnetic resonance angiography and neck vascular ultrasound examination were used to confirm the diagnosis of intracranial and carotid artery stenoses. The Mini-Mental State Examination was used to assess the cognitive function of each participant. Logistic regression was used to evaluate the relationship between tHcy levels and cognitive function. RESULTS This study included 236 participants (mean age: 64.0 (SD, 7.5) years, female: 58.1%). Multivariable analyses adjusted for several potential confounders, including creatinine and cardiovascular risk factors, showed that tHcy was associated with carotid artery stenosis (CAS). After adjusting for CAS, ICAS and several potential confounders, the association between tHcy level and low cognitive function remained significant (odds ratio: 1.09, 95% confidence interval: (1.03, 1.16), P = 0.032) . CONCLUSION Increased serum tHcy level was associated with low cognitive function independent of asymptomatic intracranial and carotid arteries stenoses.
Collapse
Affiliation(s)
- Xiaona Wang
- Department of Rehabilitation, 34708Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shangdong Province, China
| | - Tian Qiao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Min Liu
- Department of Rehabilitation, 34708Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shangdong Province, China
| | - Xiang Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.,Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
26
|
Ma H, Zhou T, Li X, Heianza Y, Qi L. Use of fish oil supplements is differently related to incidence of all-cause and vascular dementia among people with the distinct APOE ε4 dosage. Clin Nutr 2022; 41:731-736. [PMID: 35172255 PMCID: PMC8897253 DOI: 10.1016/j.clnu.2022.01.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUNDS &AIMS Previous studies have shown that marine omega-3 PUFAs (fish oil) supplements was associated with improved cognitive function, whereas the association between use of fish oil supplements and risk of incident dementia was still unclear. We aimed to prospectively assess the relations between use of fish oil supplements and risks of all-cause and disease-specific dementia according to the apolipoprotein E (APOE) ε4 dosage. METHODS A total of 445,961 participants from UK biobank, who were free of dementia at baseline and completed data on supplement use and genetic information were analyzed in this study. Cox proportional hazards models were used to calculate the hazard ratios (HRs) comparing incident dementia rates in participants who did and did not use fish oil. RESULTS During a median of 12.2 years of follow-up, a total of 5795 incident cases of dementia were documented, including 1266 cases of vascular dementia and 2382 cases of AD. After adjustment for covariates, use of fish oil supplements was significantly associated with lower risks of all-cause dementia (Hazard ratios, HR, 95% CI, 0.90, 0.85-0.96) and vascular dementia (HR, 0.85; 95% CI, 0.75-0.97), but not AD (HR, 0.99; 95% CI, 0.91-1.09). For all-cause dementia and vascular dementia, we found that the protective associations appeared to be attenuated by the increasing APOE ε4 dosage (P-interaction = 0.002 and 0.002, respectively). Notably, the use of fish oil supplements was significantly associated with an 86.0% higher risk of vascular dementia in participants with two APOE-ε4 alleles (HR, 1.86, 95%CI, 1.23-2.80). CONCLUSIONS Our results indicate that use of fish oil supplements is differently associated with risks of all-cause dementia and vascular dementia according to the APOE ε4 dosage.
Collapse
Affiliation(s)
- Hao Ma
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Tao Zhou
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
27
|
De Anda-Duran I, Alonso CF, Libon DJ, Carmichael OT, Kolachalama VB, Suglia SF, Au R, Bazzano LA. Carotid Intima-media Thickness and Midlife Cognitive Function: Impact of Race and Social Disparities in the Bogalusa Heart Study. Neurology 2022; 98:e1828-e1836. [PMID: 35228334 PMCID: PMC9109147 DOI: 10.1212/wnl.0000000000200155] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/18/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Carotid intima-media thickness (c-IMT) is a measurement of atherosclerosis, a progressive disease that develops as early as childhood and has been linked with cognitive impairment and dementia in the elderly. However, the relationship between c-IMT and midlife cognitive function as well as race and social disparities on this relationship remains unclear. We examined the association between c-IMT and cognitive function in midlife among Black and White participants from a semi-rural community-based cohort in Bogalusa, Louisiana. METHODS In this cross-sectional analysis of participants from the Bogalusa Heart Study, linear regression models were used to determine the association between c-IMT dichotomized above the 50th percentile (> 0.87mm), an a demographically standardized global cognitive (GCS) and individual cognitive domain-based z-scores. Stratified analyses were performed to evaluate the impact of race and the individual's education status. RESULTS A total of 1,217 participants (age 48 ± 5.28 years) were included, 66% (804) self-identified as White and 34% (413) as Black. Of those, 58% (708) were women, and 42% (509) were men Having a c-IMT > 50th percentile was inversely associated with GCS ([B ± SE]: -0.39 ± 0.18, P=0.03), independent of cardiovascular risk factors (CVRFs) and achieved education. The effect remained significant in Black and White participants after adjustment for CVRFs (Blacks: [B ± SE]: -1.25 ± 0.45, P=0.005; Whites: [B ± SE]: -0.92 ± 0.35, P=0.008), but not for education. The interaction between c-IMT >50th percentile and education was significant (P=0.03), and stratified analysis showed an association with GCS among those with lower achieved education ([B ± SE]: -0.81 ± 0.33, P=0.013) independent of major CVRFs. DISCUSSION Subclinical atherosclerosis, measured as c-IMT, was associated with worse midlife cognitive function, independent of major CVRFs. The association was buffered by education and may be stronger among Black than White participants, likely due to corresponding structural and social determinants. These findings underscore the importance of establishing preventive measures in midlife and suggest subclinical atherosclerosis as a potential target to prevent cognitive decline.
Collapse
Affiliation(s)
- Ileana De Anda-Duran
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | | | - David J Libon
- Department of Psychology, Rowan University, Glassboro, NJ, USA.,New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Glassboro, NJ, USA
| | - Owen T Carmichael
- Louisiana State University's Pennington Biomedical Research Centre, Baton Rouge, LA, USA
| | - Vijaya B Kolachalama
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Computer Science and Faculty of Computing and Data Sciences , Boston University, Boston, MA, USA
| | - Shakira F Suglia
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA, and Boston University Alzheimer's Disease Center, Boston, MA, USA
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
28
|
Wang W, Gottesman RF, Meyer ML, Hughes TM, Sullivan KJ, Wong DF, Lakshminarayan K, Lutsey PL. Carotid Intima-Media Thickness and Amyloid-β Deposition: The ARIC-PET Study. J Alzheimers Dis 2022; 88:17-22. [PMID: 35527548 PMCID: PMC10167843 DOI: 10.3233/jad-215671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We assessed whether carotid intima-media thickness (cIMT) is prospectively associated with amyloid-β (Aβ). 332 nondemented Atherosclerosis Risk in Communities Study participants with carotid ultrasounds (1990-1992) and PET scans (2012-2014) were studied. Participants in the highest (versus lowest) cIMT tertile had 2.17 times the odds of elevated Aβ (95% CI: 1.15-4.11), after demographic and APOE ɛ4 adjustments. An interaction with APOE ɛ4 was observed (p = 0.02). Greater cIMT was associated with elevated Aβ independent of vascular risk factors among those with ≥1 APOE ɛ4 allele, but not in noncarriers. In this cohort, higher cIMT was associated with Aβ deposition 22 years later, particularly among APOE ɛ4 carriers.
Collapse
Affiliation(s)
- Wendy Wang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Rebecca F. Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, Maryland
| | - Michelle L. Meyer
- Department of Emergency Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Timothy M. Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kevin J. Sullivan
- Department of Medicine: The MIND Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Dean F. Wong
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri
- Department of Neuroscience, Washington University in St. Louis, St. Louis, Missouri
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Kamakshi Lakshminarayan
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
29
|
Tang GL. The tangled story of carotid disease, carotid revascularization, and Alzheimer's disease: The plot thickens. J Vasc Surg 2021; 75:229. [PMID: 34949379 DOI: 10.1016/j.jvs.2021.08.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 11/18/2022]
Affiliation(s)
- Gale L Tang
- Division of Vascular Surgery, University of Washington, Seattle, WA; VA Puget Sound, Division of Vascular Surgery, Seattle, WA
| |
Collapse
|
30
|
Fresnais D, Humble MB, Bejerot S, Meehan A, Fure B. The Association between Carotid Intima-Media Thickness and Cognitive Impairment: A Systematic Review and Meta-Analysis. Dement Geriatr Cogn Disord 2021; 50:305-317. [PMID: 34808621 DOI: 10.1159/000518295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/04/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Emerging evidence suggests that cognitive impairment (CI) and different etiologies of dementia, including Alzheimer's disease (AD), are associated with vascular risk factors and atherosclerosis. In clinical practice, carotid intima-media thickness (CIMT) measured by ultrasonography may be a marker of atherosclerosis. Many studies report increased CIMT in patients with dementia and CI although a firm association has not yet been established. AIM This systematic review and meta-analysis were conducted to study the relationship between CIMT, dementia, and CI. METHODS The literature search was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines and included the following databases: Medline, Embase, Cochrane Library, and Epistemonikos. The search spanned from 2000 to 2020 and was limited to English and Scandinavian languages. RESULTS The main analysis of CIMT in subjects with CI compared to subjects with no cognitive impairment (NCI) included 12 studies; 1,089 subjects with CI and 5,223 with NCI. There was no significant difference in CIMT between the CI and NCI groups. However, subgroup analyses revealed significantly higher CIMT in the mild cognitive impairment (MCI) and dementia groups than the NCI group. In addition, patients with dementia had increased CIMT compared to patients with MCI, and patients with AD demonstrated higher CIMT than those with vascular cognitive impairment (VCI). CONCLUSION CIMT may be higher in subjects with CI than in cognitively healthy subjects although no significant difference was observed in our main analysis. CIMT was higher in the dementia group than the MCI group and in the AD group compared to the VCI group.
Collapse
Affiliation(s)
- David Fresnais
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Department of Internal Medicine, Central Hospital Karlstad, Karlstad, Sweden
| | - Mats B Humble
- School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Susanne Bejerot
- School of Medical Sciences, Örebro University, Örebro, Sweden.,University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Adrian Meehan
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Department of Geriatrics, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Brynjar Fure
- School of Medical Sciences, Örebro University, Örebro, Sweden.,Department of Internal Medicine, Central Hospital Karlstad, Karlstad, Sweden
| |
Collapse
|
31
|
The association between carotid intima-media thickness and cognitive performance is affected by intracranial artery stenosis in Chinese elderly people: An outpatient-based study. J Clin Neurosci 2021; 96:199-204. [PMID: 34840094 DOI: 10.1016/j.jocn.2021.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/14/2021] [Accepted: 11/10/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS The association between carotid intima-media thickness (cIMT) and cognitive function remains controversial, and whether this relationship is affected by intracranial artery stenosis (ICAS) remains unclear. We investigated these questions among elderly participants who sought health consultations in an outpatient clinic. METHODS We conducted a cross-sectional study based on participants from an outpatient clinic, enrolling residents over 60 years of age seeking outpatient services because of abnormal transcranial Doppler reports at Shandong Provincial Third Hospital in Jinan, Shandong province. We performed physical examinations, blood tests, cIMT measurement using carotid ultrasonography, ICAS measurement using brain magnetic resonance angiography scanning, and global cognitive function assessment using the Montreal Cognitive Assessment (MoCA)in the outpatient clinic from May 2020 to December 2020. We subsequently performed a regression analysis to explore the relationship between cIMT and cognitive function and a stratified analysis to explore whether the relationship was different between the ICAS and non-ICAS participants. RESULTS In total, 167 participants (age: 65.56 ± 10.39 years, female: 53.89%) were included in the present study. The MoCA score was significantly lower in the intimal thickening group (cIMT ≥ 1.0 mm) than in the normal group (mean [SD]: 16.23 [5.16] vs. 19.97 [4.59], P < 0.001). Univariate analysis showed that a greater cIMT was negatively correlated with cognitive function. After adjustment for several potential confounders in the multivariable analyses, the association between cIMT and cognitive function disappeared. When further stratified by ICAS, a negative association between increased cIMT (cIMT ≥ 1.0 mm) and cognitive function was found in those without ICAS (β: -2.80 [-5.13, -0.48], p = 0.021); however, in subjects with ICAS, the relationship between cIMT and cognitive function was insignificant. CONCLUSION Greater cIMT was associated with low cognitive function in participants without ICAS who sought consultation due to abnormal transcranial Doppler reports in outpatient clinics.
Collapse
|
32
|
Claassen JAHR, Thijssen DHJ, Panerai RB, Faraci FM. Regulation of cerebral blood flow in humans: physiology and clinical implications of autoregulation. Physiol Rev 2021; 101:1487-1559. [PMID: 33769101 PMCID: PMC8576366 DOI: 10.1152/physrev.00022.2020] [Citation(s) in RCA: 339] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain function critically depends on a close matching between metabolic demands, appropriate delivery of oxygen and nutrients, and removal of cellular waste. This matching requires continuous regulation of cerebral blood flow (CBF), which can be categorized into four broad topics: 1) autoregulation, which describes the response of the cerebrovasculature to changes in perfusion pressure; 2) vascular reactivity to vasoactive stimuli [including carbon dioxide (CO2)]; 3) neurovascular coupling (NVC), i.e., the CBF response to local changes in neural activity (often standardized cognitive stimuli in humans); and 4) endothelium-dependent responses. This review focuses primarily on autoregulation and its clinical implications. To place autoregulation in a more precise context, and to better understand integrated approaches in the cerebral circulation, we also briefly address reactivity to CO2 and NVC. In addition to our focus on effects of perfusion pressure (or blood pressure), we describe the impact of select stimuli on regulation of CBF (i.e., arterial blood gases, cerebral metabolism, neural mechanisms, and specific vascular cells), the interrelationships between these stimuli, and implications for regulation of CBF at the level of large arteries and the microcirculation. We review clinical implications of autoregulation in aging, hypertension, stroke, mild cognitive impairment, anesthesia, and dementias. Finally, we discuss autoregulation in the context of common daily physiological challenges, including changes in posture (e.g., orthostatic hypotension, syncope) and physical activity.
Collapse
Affiliation(s)
- Jurgen A H R Claassen
- Department of Geriatrics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen, The Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Ronney B Panerai
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- >National Institute for Health Research Leicester Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Frank M Faraci
- Departments of Internal Medicine, Neuroscience, and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
33
|
Huang YT, Hong FF, Yang SL. Atherosclerosis: The Culprit and Co-victim of Vascular Dementia. Front Neurosci 2021; 15:673440. [PMID: 34421513 PMCID: PMC8377286 DOI: 10.3389/fnins.2021.673440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/11/2021] [Indexed: 11/24/2022] Open
Abstract
Vascular dementia (VD), a cerebrovascular disease which causes cognitive impairment, is one of the significant factors that affects the quality of senectitude. Atherosclerosis (AS) is a chronic inflammatory syndrome and closely associated with VD. Analyzing the role of AS in VD contribute greatly to its early detection and prevention, but their relationship has not been integrated into a complete network. This review summarizes AS biomarkers as VD predictors for the first time and describes the direct mechanisms of AS causing VD from five aspects: vascular morphogenesis, hemodynamic change, neurovascular unit damage (NVU), oxidative stress, and microRNA (miRNA). Finally, it discriminates the relationship between AS and VD in common risk factors which can be disease or some molecules. In particular, these data imply that the role of AS in VD is not only a pathogenic factor but also a comorbidity in VD. This review aims to bring new ideas for the prediction and treatment of VD.
Collapse
Affiliation(s)
- Ya-Ting Huang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.,Queen Marry College, School of Medicine, Nanchang University, Nanchang, China
| | - Fen-Fang Hong
- Experimental Center of Pathogen Biology, Nanchang University, Nanchang, China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, China.,Department of Physiology, Fuzhou Medical College, Fuzhou, China
| |
Collapse
|
34
|
Pinho J, Quintas-Neves M, Dogan I, Reetz K, Reich A, Costa AS. Incident stroke in patients with Alzheimer's disease: systematic review and meta-analysis. Sci Rep 2021; 11:16385. [PMID: 34385535 PMCID: PMC8361108 DOI: 10.1038/s41598-021-95821-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/29/2021] [Indexed: 11/09/2022] Open
Abstract
Vascular mechanisms are increasingly recognized in the pathophysiology of Alzheimer's disease (AD), but less is known about the occurrence of stroke in AD patients. We aimed to quantify the risk of stroke in patients with AD and compare the incidence rates (IR) of stroke in individuals without AD. Systematic search of Embase and MEDLINE between 1970 and 2020. Inclusion criteria: reports with ≥ 50 patients with non-familial AD, which reported the occurrence of stroke (all types) and/or ischemic stroke and/or intracerebral hemorrhage (ICH) during follow-up. Meta-analyses of pooled data using random-effects model were performed. IR were calculated for each study. Incidence rate ratios (IRR) were calculated for studies presenting a control-group without AD. Among 5109 retrieved studies, 29 (0.6%) fulfilled the inclusion criteria, reporting a total of 61,824 AD patients. In AD patients the IR were 15.4/1000 person-years for stroke (all types), 13.0/1000 person-years for ischemic stroke and 3.4/1000 person-years for ICH. When compared to controls without AD, incidence rate for ICH in AD patients was significantly higher (IRR = 1.67, 95%CI 1.43-1.96), but similar for ischemic stroke. Incident stroke is not a rare event in AD population. AD is associated with an increased risk of intracerebral hemorrhage which warrants further clarification.
Collapse
Affiliation(s)
- João Pinho
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsst. 30, 52074, Aachen, Germany.
| | - Miguel Quintas-Neves
- Neuroradiology Department, Hospital de Braga, Braga, Portugal.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B´s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Imis Dogan
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsst. 30, 52074, Aachen, Germany.,JARA Institute Molecular Neuroscience and Neuroimaging, Juelich Research Center GmbH and RWTH Aachen University, Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsst. 30, 52074, Aachen, Germany.,JARA Institute Molecular Neuroscience and Neuroimaging, Juelich Research Center GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsst. 30, 52074, Aachen, Germany
| | - Ana Sofia Costa
- Department of Neurology, University Hospital RWTH Aachen, Pauwelsst. 30, 52074, Aachen, Germany.,JARA Institute Molecular Neuroscience and Neuroimaging, Juelich Research Center GmbH and RWTH Aachen University, Aachen, Germany
| |
Collapse
|
35
|
Ihle-Hansen H, Ihle-Hansen H, Sandset EC, Hagberg G. Subclinical Carotid Artery Atherosclerosis and Cognitive Function: A Mini-Review. Front Neurol 2021; 12:705043. [PMID: 34393982 PMCID: PMC8355501 DOI: 10.3389/fneur.2021.705043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022] Open
Abstract
Carotid artery atherosclerosis, the result of a multitude of vascular risk factors, is a promising marker for use in risk stratification. Recent evidence suggests that carotid artery atherosclerosis affects cognitive function and is an independent risk factor for the development of cognitive impairment. Both atherosclerosis and cognitive impairment develop over a prolonged period (years), and due to the aging population, markers to identify persons at risk are needed. Carotid artery atherosclerosis can easily be visualized using non-invasive ultrasound, potentially enabling early and intensified risk factor management to preserve cognitive function or delay further decline. However, the burden of atherosclerosis and temporal exposure required to pose a risk of cognitive impairment is unclear. This mini-review aims to explore the available evidence on the association between carotid atherosclerosis and cognition, and furthermore identify the remaining gaps in knowledge.
Collapse
Affiliation(s)
- Håkon Ihle-Hansen
- Department of Medicine, Bærum Hospital- Vestre Viken Hospital Trust, Drammen, Norway
| | - Hege Ihle-Hansen
- Department of Medicine, Bærum Hospital- Vestre Viken Hospital Trust, Drammen, Norway.,Oslo Stroke Unit, Department of Neurology, Oslo University Hospital, Ullevål, Norway
| | | | - Guri Hagberg
- Department of Medicine, Bærum Hospital- Vestre Viken Hospital Trust, Drammen, Norway.,Oslo Stroke Unit, Department of Neurology, Oslo University Hospital, Ullevål, Norway
| |
Collapse
|
36
|
Wu X, Liu Y, Zhu L, Wang Y, Ren Y, Cheng B, Ren L, Ge K, Li H. Cerebroprotein Hydrolysate-I Inhibits Hippocampal Neuronal Apoptosis by Activating PI3K/Akt Signaling Pathway in Vascular Dementia Mice. Neuropsychiatr Dis Treat 2021; 17:2359-2368. [PMID: 34305399 PMCID: PMC8297406 DOI: 10.2147/ndt.s311760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Vascular dementia (VaD), one of the brain injuries, is difficult to be cured, so it is important to take active neuroprotective treatment after its occurrence. Many studies have shown that apoptosis serves an important role in VaD occurrence; therefore, inhibition of apoptosis may contribute to the recovery of neurological function after VaD occurrence. Cerebroprotein hydrolysate-I (CH-I), a neuropeptide preparation which consists of several amino acids and small molecular peptides as the main active constituent, is extracted using a method similar to cerebrolysin (CBL) which has neuroprotective and neurotrophic effects. METHODS In the present study, a VaD model which was constructed using bilateral common carotid artery occlusion (BCCAO) in Kunming mice was applied to examine the neuroprotective effects of CH-I. RESULTS The results show that CH-I treatment could attenuate the decrease of learning and memory ability, cell apoptosis in the hippocampal CA1 region and inhibit the activation of caspase-3 and caspase-9 in VaD mice. Furthermore, CH-I treatment could also upregulate Bcl-2 protein levels and activate PI3K and Akt. DISCUSSION We speculate that CH-I may induce a neuroprotective effect activating PI3K/Akt signaling pathway in VaD mice.
Collapse
Affiliation(s)
- Xiaolin Wu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, People’s Republic of China
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Yingjuan Liu
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Lin Zhu
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Yue Wang
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Yuqian Ren
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Baohe Cheng
- Shandong Haoyun Guoji Stem Cells Hospital, Jinan, Shandong, 250001, People’s Republic of China
| | - Leiming Ren
- Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei, 050017, People’s Republic of China
| | - Keli Ge
- Institute of Integrative Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong, 266021, People’s Republic of China
| | - Hongyun Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, People’s Republic of China
| |
Collapse
|
37
|
Bir SC, Khan MW, Javalkar V, Toledo EG, Kelley RE. Emerging Concepts in Vascular Dementia: A Review. J Stroke Cerebrovasc Dis 2021; 30:105864. [PMID: 34062312 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105864] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Vascular dementia (VaD) is the second most common cause of dementia and a major health concern worldwide. A comprehensive review on VaD is warranted for better understanding and guidance for the practitioner. We provide an updated overview of the epidemiology, pathophysiological mechanisms, neuroimaging patterns as well as current diagnostic and therapeutic approaches. MATERIALS AND METHODS A narrative review of current literature in VaD was performed based on publications from the database of PubMed, Scopus and Google Scholar up to January, 2021. RESULTS VaD can be the result of ischemic or hemorrhagic tissue injury in a particular region of the brain which translates into clinically significant cognitive impairment. For example, a cerebral infarct in the speech area of the dominant hemisphere would translate into clinically significant impairment as would involvement of projection pathways such as the arcuate fasciculus. Specific involvement of the angular gyrus of the dominant hemisphere, with resultant Gerstman's syndrome, could have a pronounced effect on functional ability despite being termed a "minor stroke". Small vessel cerebrovascular disease can have a cumulate effect on cognitive function over time. It is unfortunately well recognized that "good" functional recovery in acute ischemic or haemorrhagic stroke, including subarachnoid haemorrhage, does not necessarily translate into good cognitive recovery. The victim may often be left unable to have gainful employment, drive a car safely or handle their affairs independently. CONCLUSIONS This review should serve as a compendium of updated information on VaD and provide guidance in terms of newer diagnostic and potential therapeutic approaches.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Muhammad W Khan
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Vijayakumar Javalkar
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | | | - Roger E Kelley
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA.
| |
Collapse
|
38
|
Scheltens P, De Strooper B, Kivipelto M, Holstege H, Chételat G, Teunissen CE, Cummings J, van der Flier WM. Alzheimer's disease. Lancet 2021; 397:1577-1590. [PMID: 33667416 PMCID: PMC8354300 DOI: 10.1016/s0140-6736(20)32205-4] [Citation(s) in RCA: 1781] [Impact Index Per Article: 593.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 08/21/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022]
Abstract
In this Seminar, we highlight the main developments in the field of Alzheimer's disease. The most recent data indicate that, by 2050, the prevalence of dementia will double in Europe and triple worldwide, and that estimate is 3 times higher when based on a biological (rather than clinical) definition of Alzheimer's disease. The earliest phase of Alzheimer's disease (cellular phase) happens in parallel with accumulating amyloid β, inducing the spread of tau pathology. The risk of Alzheimer's disease is 60-80% dependent on heritable factors, with more than 40 Alzheimer's disease-associated genetic risk loci already identified, of which the APOE alleles have the strongest association with the disease. Novel biomarkers include PET scans and plasma assays for amyloid β and phosphorylated tau, which show great promise for clinical and research use. Multidomain lifestyle-based prevention trials suggest cognitive benefits in participants with increased risk of dementia. Lifestyle factors do not directly affect Alzheimer's disease pathology, but can still contribute to a positive outcome in individuals with Alzheimer's disease. Promising pharmacological treatments are poised at advanced stages of clinical trials and include anti-amyloid β, anti-tau, and anti-inflammatory strategies.
Collapse
Affiliation(s)
- Philip Scheltens
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Life Science Partners, Amsterdam, Netherlands.
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurology, Leuven, Belgium; Dementia Research Institute, University College London, London, UK
| | - Miia Kivipelto
- Division of Clinical Geriatrics and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska University Hospital, Stockholm, Sweden; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Ageing and Epidemiology Research Unit, School of Public Health, Imperial College London, London, UK
| | - Henne Holstege
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Gael Chételat
- Normandie Université, Université de Caen, Institut National de la Santé et de la Recherche Médicale, Groupement d'Intérêt Public Cyceron, Caen, France
| | - Charlotte E Teunissen
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, University of Nevada, Las Vegas, NV, USA; Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Wiesje M van der Flier
- Alzheimer Centre Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Epidemiology and Datascience, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
39
|
Vinke EJ, Yilmaz P, van der Toorn JE, Fakhry R, Frenzen K, Dubost F, Licher S, de Bruijne M, Kavousi M, Ikram MA, Vernooij MW, Bos D. Intracranial arteriosclerosis is related to cerebral small vessel disease: a prospective cohort study. Neurobiol Aging 2021; 105:16-24. [PMID: 34004492 DOI: 10.1016/j.neurobiolaging.2021.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 11/16/2022]
Abstract
Intracranial arteriosclerosis has been increasingly recognized as a risk factor for cognitive impairment and even dementia. A possible mechanism linking intracranial arteriosclerosis to cognitive impairment and dementia involves structural brain changes including cerebral small vessel disease (CSVD). To assess whether intracranial carotid artery calcification (ICAC) and vertebrobasilar artery calcification (VBAC), as proxies for intracranial arteriosclerosis, are related to CSVD. Within the population-based Rotterdam Study, between 2003 and 2006 a computed tomography (CT)-based measurement of ICAC and VBAC and at least one magnetic resonance imaging (MRI) measurement of structural brain changes were performed from 2005 onwards in 1,489 participants. To estimate the burden of calcification independent of age, we computed age-adjusted percentile curves for ICAC and VBAC separately, based on the calcification volumes. Using the longitudinal MRI data, we assessed whether a larger calcification burden accelerates structural brain changes using appropriate statistical models for repeated outcome measures. A larger burden of ICAC and VBAC was associated with an increase of CSVD markers accelerating over time. A larger burden of ICAC and VBAC was not significantly (p > 0.05) associated with accelerated brain atrophy. Arteriosclerosis is related to accelerating structural brain changes over time.
Collapse
Affiliation(s)
- Elisabeth J Vinke
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Pinar Yilmaz
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Janine E van der Toorn
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rahman Fakhry
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Kate Frenzen
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Florian Dubost
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Silvan Licher
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Marleen de Bruijne
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Daniel Bos
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands; Department of Clinical Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
40
|
Cortes-Canteli M, Gispert JD, Salvadó G, Toribio-Fernandez R, Tristão-Pereira C, Falcon C, Oliva B, Mendiguren J, Fernandez-Friera L, Sanz J, Garcia-Ruiz JM, Fernandez-Ortiz A, Sanchez-Gonzalez J, Ibanez B, Molinuevo JL, Fuster V. Subclinical Atherosclerosis and Brain Metabolism in Middle-Aged Individuals: The PESA Study. J Am Coll Cardiol 2021; 77:888-898. [PMID: 33602472 DOI: 10.1016/j.jacc.2020.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atherosclerosis has been linked to cognitive decline in late life; however, the impact of cardiovascular risk factors (CVRFs) and subclinical atherosclerosis on brain metabolism at earlier stages remains unexplored. OBJECTIVES This study sought to determine the association between brain metabolism, subclinical atherosclerosis, and CVRFs in middle-aged asymptomatic individuals. METHODS This study included 547 asymptomatic middle-aged participants (50 ± 4 years, 82% men) from the PESA (Progression of Early Subclinical Atherosclerosis) study with evidence of subclinical atherosclerosis. Participants underwent 18F-fluorodeoxyglucose (FDG)-positron emission tomography. Global brain FDG uptake and voxel-wise analyses were used to evaluate the associations of cerebral metabolism with CVRFs and atherosclerotic plaque burden in carotids and femorals assessed by 3-dimensional vascular ultrasound. RESULTS Global FDG uptake showed an inverse correlation with 30-year Framingham Risk Score (FRS) (β = -0.15, p < 0.001). This association was mainly driven by the presence of hypertension (d = 0.36, p < 0.001). Carotid plaque burden was inversely associated with global brain FDG uptake (β = -0.16, p < 0.001), even after adjusting for 30-year FRS. Voxel-wise approaches revealed that the brain areas most strongly affected by hypometabolism in association with 30-year FRS, hypertension, and carotid plaque burden were parietotemporal regions (angular, supramarginal, and inferior/middle temporal gyri) and the cingulate gyrus. CONCLUSIONS In asymptomatic middle-aged individuals, cardiovascular risk is associated with brain hypometabolism, with hypertension being the modifiable CVRF showing the strongest association. Subclinical carotid plaque burden is also linked to reduced brain metabolism independently of CVRFs. Cerebral areas showing hypometabolism include those known to be affected in dementia. These data reinforce the need to control CVRFs early in life in order to potentially reduce the brain's midlife vulnerability to future cognitive dysfunction.
Collapse
Affiliation(s)
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Belen Oliva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Leticia Fernandez-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; HM Hospitales-Centro Integral de Enfermedades Cardiovasculares, Universidad San Pablo-CEU, Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain
| | - Javier Sanz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jose M Garcia-Ruiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Universitario Central de Oviedo, Asturias, Spain
| | - Antonio Fernandez-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos, Universidad Complutense, Madrid, Spain
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Madrid, Spain.
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
41
|
Liu L, Gracely EJ, Yin X, Eisen HJ. Impact of diabetes mellitus and cardiometabolic syndrome on the risk of Alzheimer’s disease among postmenopausal women. World J Diabetes 2021; 12:69-83. [PMID: 33520109 PMCID: PMC7807256 DOI: 10.4239/wjd.v12.i1.69] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/13/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In spite of an increase in the incidence and prevalence of diabetes mellitus (DM) and Alzheimer’s disease (AD) in the aging population worldwide, limited attention has been paid to their potential association.
AIM To investigate the association of DM and cardiometabolic syndrome (CMS, a precursor to DM) with risk of incident AD among postmenopausal women.
METHODS Postmenopausal women aged 50-79 (n = 63117) who participated in the U.S. Women’s Health Initiative Observational Study (WHIOS), recruited in 1993-1998, without baseline AD and followed up through March 1, 2019, were analyzed. AD was classified by participant-reported history of doctor-diagnosis of incident AD in the WHIOS. DM was defined by participant-report or treated because of diabetes or serum glucose concentrations ≥ 126 mg/dL. CMS was defined as having ≥ 3 of five CMS components: large waist circumference, high blood pressure, elevated triglycerides, elevated glucose, and low high-density lipoprotein cholesterol. The associations of DM and CMS with AD were analyzed using Cox’s proportional hazards regression analysis.
RESULTS During a median follow-up of 20 years (range: 3.36 to 23.36 years), of 63117 participants, 8340 developed incident AD. Women with DM had significantly higher incidence of AD [8.5, 95% confidence interval (CI): 8.0-9.0 per 1000 person-years (PY)] than those without DM (7.1, 95%CI: 6.9-7.2 per 1000 PY). Multivariate Cox’s regression analysis indicated that women with DM or CMS had a significantly higher risk of AD than those without DM or CMS. The corresponding hazard ratios [HR (95%CI)] were 1.22 (1.13-1.31, P < 0.001) in subjects with DM, and 1.18 (1.09-1.27, P < 0.001) in subjects with CMS. The HRs diminished with age and became non-significant in the oldest age group.
CONCLUSION During a median follow-up of 20 years, DM and CMS were significantly associated with the risk of AD among postmenopausal women. More specifically, women aged 50-69 with DM or CMS vs those without these conditions had significantly higher relative risks of AD than the relative risks of AD in those aged 70-79 with DM or CMS vs those without DM or CMS.
Collapse
Affiliation(s)
- Longjian Liu
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, United States
| | - Edward J Gracely
- Department of Epidemiology and Biostatistics, Dornsife School of Public Health, Drexel University, Philadelphia, PA 19104, United States
- Department of Family, Community and Preventive Medicine, Drexel University College of Medicine, Philadelphia, PA 19129, United States
| | - Xiaoyan Yin
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Howard J Eisen
- Heart and Vascular Institute, Pennsylvania State University, Hershey, PA 17033, United States
| |
Collapse
|
42
|
Affiliation(s)
- Lewis H Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
43
|
Byman E, Nägga K, Gustavsson AM, Andersson-Assarsson J, Hansson O, Sonestedt E, Wennström M. Alpha-amylase 1A copy number variants and the association with memory performance and Alzheimer's dementia. ALZHEIMERS RESEARCH & THERAPY 2020; 12:158. [PMID: 33220711 PMCID: PMC7680592 DOI: 10.1186/s13195-020-00726-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022]
Abstract
Background Previous studies have shown that copy number variation (CNV) in the alpha (α)-amylase gene (AMY1A) is associated with body mass index, insulin resistance, and blood glucose levels, factors also shown to increase the risk of Alzheimer’s dementia (AD). We have previously demonstrated the presence of α-amylase in healthy neuronal dendritic spines and a reduction of the same in AD patients. In the current study, we investigate the relationship between AMY1A copy number and AD, memory performance, and brain α-amylase activity. Methods and materials The association between AMY1A copy number and development of AD was analyzed in 5422 individuals (mean age at baseline 57.5 ± 5.9, females 58.2%) from the Malmö diet and cancer study genotyped for AMY1A copy number, whereof 247 where diagnosed with AD during a mean follow-up of 20 years. Associations between AMY1A copy number and cognitive performance where analyzed in 791 individuals (mean age at baseline 54.7 ± 6.3, females 63%), who performed Montreal Cognitive Assessment (MoCA) test. Correlation analysis between α-amylase activity or α-amylase gene expression and AMY1A copy number in post-mortem hippocampal tissue from on demented controls (n = 8) and AD patients (n = 10) was also performed. Results Individuals with very high ( ≥10) AMY1A copy number had a significantly lower hazard ratio of AD (HR = 0.62, 95% CI 0.41–0.94) and performed significantly better on MoCA delayed word recall test, compared to the reference group with AMY1A copy number 6. A trend to lower hazard ratio of AD was also found among individuals with low AMY1A copy number (1–5) (HR = 0.74, 95% CI 0.53–1.02). A tendency towards a positive correlation between brain α-amylase activity and AMY1A copy number was found, and females showed higher brain α-amylase activity compared to males. Conclusion Our study suggests that the degree of α-amylase activity in the brain is affected by AMY1A copy number and gender, in addition to AD pathology. The study further suggests that very high AMY1A copy number is associated with a decreased hazard ratio of AD and we speculate that this effect is mediated via a beneficial impact of AMY1A copy number on episodic memory performance.
Collapse
Affiliation(s)
- Elin Byman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Inga Marie Nilssons gata 53, SE-214 28, Malmö, Sweden
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Inga Marie Nilssons gata 53, SE-214 28, Malmö, Sweden.,Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Anna-Märta Gustavsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Inga Marie Nilssons gata 53, SE-214 28, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | | | - Johanna Andersson-Assarsson
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Inga Marie Nilssons gata 53, SE-214 28, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Emily Sonestedt
- Nutritional Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Malin Wennström
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Inga Marie Nilssons gata 53, SE-214 28, Malmö, Sweden.
| |
Collapse
|
44
|
Doorduijn AS, de van der Schueren MAE, van de Rest O, de Leeuw FA, Hendriksen HMA, Teunissen CE, Scheltens P, van der Flier WM, Visser M. Nutritional Status Is Associated With Clinical Progression in Alzheimer's Disease: The NUDAD Project. J Am Med Dir Assoc 2020; 24:638-644.e1. [PMID: 33239240 DOI: 10.1016/j.jamda.2020.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/21/2020] [Accepted: 10/10/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In cognitively normal adults, nutritional parameters are related to cognitive decline and incidence of dementia. Studies on the role of nutrition in predementia stages subjective cognitive decline and mild cognitive impairment, and mild stages of Alzheimer's disease (AD) dementia in a clinical setting are lacking. In the absence of a curative treatment, this evidence is important for targeting nutritional factors to potentially prevent or delay further cognitive decline. Our aim is to investigate associations of nutritional parameters with clinical progression in patients ranging from those who are cognitively normal to those who have AD dementia. DESIGN Longitudinal. SETTING AND PARTICIPANTS Memory clinic, 551 patients (219 with subjective cognitive decline, 135 with mild cognitive impairment, and 197 with AD dementia), mean age 64 ± 8 years. MEASUREMENTS We assessed body mass index, fat-free mass, Mini-Nutritional Assessment, and dietary intake with the Dutch Healthy Diet food frequency questionnaire and the 238-item healthy life in an urban setting (HELIUS) food frequency questionnaire at baseline. Cox proportional hazard models were used to evaluate associations of nutritional parameters with clinical progression. Additional analyses were restricted to patients who were amyloid positive. RESULTS We observed clinical progression in 170 patients (31%) over 2.2 ± 0.9 years. Poorer Mini-Nutritional Assessment score [hazard ratio (95% confidence interval) 1.39 (1.18-1.64)], lower body mass index [1.15 (0.96-1.38)], lower fat-free mass [1.40 (0.93-2.10)], and a less healthy dietary pattern [1.22 (1.01-1.48)] were associated with a higher risk of clinical progression. Similar effect sizes were found in patients who were amyloid positive. CONCLUSIONS AND IMPLICATIONS Poorer nutritional status and a less healthy dietary pattern are associated with a higher risk of clinical progression. This study provides support for investigating whether improving nutritional status can alter the clinical trajectory of AD.
Collapse
Affiliation(s)
- Astrid S Doorduijn
- Department of Nutrition and Dietetics, Amsterdam Public Health research institute, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Marian A E de van der Schueren
- Department of Nutrition and Health, School of Allied Health, HAN University of Applied Sciences, Nijmegen, the Netherlands; Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Ondine van de Rest
- Division of Human Nutrition and Health, Wageningen University and Research, Wageningen, the Netherlands
| | - Francisca A de Leeuw
- Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Heleen M A Hendriksen
- Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Philip Scheltens
- Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wiesje M van der Flier
- Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Epidemiology & Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Marjolein Visser
- Faculty of Science, Department of Health Sciences, Vrije Universiteit Amsterdam and the Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
45
|
Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, Hughes TM, Daemen MJ, Williamson JD, González HM, Schneider J, Wellington CL, Katusic ZS, Stoeckel L, Koenig JI, Corriveau RA, Fine L, Galis ZS, Reis J, Wright JD, Chen J. Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement 2020; 16:1714-1733. [PMID: 33030307 DOI: 10.1002/alz.12157] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are characterized by the aging neurovascular unit being confronted with and failing to cope with biological insults due to systemic and cerebral vascular disease, proteinopathy including Alzheimer's biology, metabolic disease, or immune response, resulting in cognitive decline. This report summarizes the discussion and recommendations from a working group convened by the National Heart, Lung, and Blood Institute and the National Institute of Neurological Disorders and Stroke to evaluate the state of the field in VCID research, identify research priorities, and foster collaborations. As discussed in this report, advances in understanding the biological mechanisms of VCID across the wide spectrum of pathologies, chronic systemic comorbidities, and other risk factors may lead to potential prevention and new treatment strategies to decrease the burden of dementia. Better understanding of the social determinants of health that affect risks for both vascular disease and VCID could provide insight into strategies to reduce racial and ethnic disparities in VCID.
Collapse
Affiliation(s)
| | | | | | - Sudha Seshadri
- University of Texas Health Science Center, San Antonio and Boston University, San Antonio, Texas, USA
| | - Ann McKee
- VA Boston Healthcare System and Boston University, Boston, Massachusetts, USA
| | | | - Steven M Greenberg
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristine Yaffe
- University of California, San Francisco, San Francisco, California, USA
| | | | - Chun Yuan
- University of Washington, Seattle, Washington, USA
| | - Timothy M Hughes
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mat J Daemen
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Luke Stoeckel
- National Institute on Aging, Bethesda, Maryland, USA
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Roderick A Corriveau
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Lawrence Fine
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Zorina S Galis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jared Reis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | | | - Jue Chen
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
46
|
Pettigrew C, Soldan A, Wang J, Wang MC, Arthur K, Moghekar A, Gottesman RF, Albert M. Association of midlife vascular risk and AD biomarkers with subsequent cognitive decline. Neurology 2020; 95:e3093-e3103. [PMID: 32989109 DOI: 10.1212/wnl.0000000000010946] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/22/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether vascular risk and Alzheimer disease (AD) biomarkers have independent or synergistic effects on cognitive decline and whether vascular risk is associated with the accumulation of AD pathology as measured by change in biomarkers over time. METHODS At baseline, participants (n = 168) were cognitively normal and primarily middle-aged (mean 56.4 years, SD 10.9 years) and had both vascular risk factor status and proximal CSF biomarkers available. Baseline vascular risk was quantified with a composite vascular risk score reflecting the presence or absence of hypertension, hypercholesterolemia, diabetes, current smoking, and obesity. CSF biomarkers of β-amyloid (Aβ)1-42, total tau (t-tau), and phosphorylated tau (p-tau) were used to create dichotomous high and low AD biomarker groups (based on Aβ1-42 and tau). Linear mixed-effects models were used to examine change in a cognitive composite score (mean follow-up 13.9 years) and change in CSF biomarkers (mean follow-up 4.2 years). RESULTS There was no evidence of a synergistic relationship between the vascular risk score and CSF AD biomarkers and cognitive decline. Instead, the vascular risk score (estimate -0.022, 95% confidence interval [CI] -0.043 to -0.002, p = 0.03) and AD biomarkers (estimate -0.060, 95% CI -0.096 to -0.024, p = 0.001) were independently and additively associated with cognitive decline. In addition, the vascular risk score was unrelated to levels of or rate of change in CSF Aβ1-42, t-tau, or p-tau. CONCLUSIONS The results of this observational cohort study suggest that vascular risk and biomarkers of AD pathology, when measured in midlife, act along independent pathways and underscore the importance of accounting for multiple risk factors for identifying cognitively normal individuals at the greatest risk of cognitive decline.
Collapse
Affiliation(s)
- Corinne Pettigrew
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD.
| | - Anja Soldan
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jiangxia Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mei-Cheng Wang
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Karissa Arthur
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Abhay Moghekar
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Rebecca F Gottesman
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Marilyn Albert
- From the Department of Neurology (C.P., A.S., K.A., A.M., R.F.G., M.A.), The Johns Hopkins University School of Medicine; and Department of Biostatistics (J.W., M.-C.W.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
47
|
Song JW, Wasserman BA. Vessel wall MR imaging of intracranial atherosclerosis. Cardiovasc Diagn Ther 2020; 10:982-993. [PMID: 32968655 DOI: 10.21037/cdt-20-470] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Intracranial atherosclerotic disease (ICAD) is one of the most common causes of ischemic stroke worldwide. Along with high recurrent stroke risk from ICAD, its association with cognitive decline and dementia leads to a substantial decrease in quality of life and a high economic burden. Atherosclerotic lesions can range from slight wall thickening with plaques that are angiographically occult to severely stenotic lesions. Recent advances in intracranial high resolution vessel wall MR (VW-MR) imaging have enabled imaging beyond the lumen to characterize the vessel wall and its pathology. This technique has opened new avenues of research for identifying vulnerable plaque in the setting of acute ischemic stroke as well as assessing ICAD burden and its associations with its sequela, such as dementia. We now understand more about the intracranial arterial wall, its ability to remodel with disease and how we can use VW-MR to identify angiographically occult lesions and assess medical treatment responses, for example, to statin therapy. Our growing understanding of ICAD with intracranial VW-MR imaging can profoundly impact diagnosis, therapy, and prognosis for ischemic stroke with the possibility of lesion-based risk models to tailor and personalize treatment. In this review, we discuss the advantages of intracranial VW-MR imaging for ICAD, the potential of bioimaging markers to identify vulnerable intracranial plaque, and future directions of artificial intelligence and its utility for lesion scoring and assessment.
Collapse
Affiliation(s)
- Jae W Song
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
48
|
Soldan A, Pettigrew C, Zhu Y, Wang M, Gottesman RF, DeCarli C, Albert M. Cognitive reserve and midlife vascular risk: Cognitive and clinical outcomes. Ann Clin Transl Neurol 2020; 7:1307-1317. [PMID: 32856790 PMCID: PMC7448143 DOI: 10.1002/acn3.51120] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Examine whether cognitive reserve moderates the association of 1) vascular risk factors and 2) white matter hyperintensity burden with risk of clinical progression and longitudinal cognitive decline. METHODS BIOCARD Study participants were cognitively normal and primarily middle-aged (M = 57 years) at baseline and have been followed with annual cognitive and clinical assessments (M = 13 years). Baseline cognitive reserve was indexed with a composite score combining education with reading and vocabulary scores. Baseline vascular risk (N = 229) was assessed with a composite risk score reflecting five vascular risk factors. Baseline white matter hyperintensity load (N = 271) was measured with FLAIR magnetic resonance imaging. Cox regression models assessed risk of progression from normal cognition to onset of clinical symptoms of Mild Cognitive Impairment. Longitudinal mixed effects models measured the relationship of these variables to cognitive decline, using a global composite score, and executive function and episodic memory sub-scores. RESULTS Both vascular risk and white matter hyperintensities were associated with cognitive decline, particularly in executive function. Higher vascular risk, but not white matter hyperintensity burden, was associated with an increased risk of progression to Mild Cognitive Impairment. Higher cognitive reserve was associated with a reduced risk of symptom onset and higher levels of baseline cognition but did not modify the associations between the vascular risk score and white matter hyperintensities with clinical progression or cognitive decline. INTERPRETATION Although cognitive reserve has protective effects on clinical and cognitive outcomes, it does not mitigate the negative impact of vascular risk and small vessel cerebrovascular disease on these same outcomes.
Collapse
Affiliation(s)
- Anja Soldan
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMD21205
| | - Corinne Pettigrew
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMD21205
| | - Yuxin Zhu
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21287
| | - Mei‐Cheng Wang
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21287
| | - Rebecca F. Gottesman
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMD21205
| | - Charles DeCarli
- Department of NeurologyUniversity of California, Davis, School of MedicineDavisCA95616
| | - Marilyn Albert
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMD21205
| | | |
Collapse
|
49
|
Abstract
Hardening of the arteries (atherosclerosis) was linked to dementia long ago, but subsequently, Alzheimer’s plaques and tangles have received more attention. A new proteome-wide association study unveils molecular links between intracranial atherosclerosis and dementia, independent of other pathologies, providing new evidence for one of the oldest suspected causes of dementia.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA.
| |
Collapse
|
50
|
Shared proteomic effects of cerebral atherosclerosis and Alzheimer's disease on the human brain. Nat Neurosci 2020; 23:696-700. [PMID: 32424284 PMCID: PMC7269838 DOI: 10.1038/s41593-020-0635-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/01/2020] [Indexed: 01/07/2023]
Abstract
Cerebral atherosclerosis contributes to dementia via unclear processes. We performed proteomic sequencing of dorsolateral prefrontal cortex in 438 older individuals and found associations between cerebral atherosclerosis and reduced synaptic signaling and RNA splicing and increased oligodendrocyte development and myelination. Consistently, single-cell RNA sequencing showed cerebral atherosclerosis associated with higher oligodendrocyte abundance. A subset of proteins and modules associated with cerebral atherosclerosis was also associated with Alzheimer’s disease, suggesting shared mechanisms.
Collapse
|