1
|
Tang H, Zhu W, Jing J, Zhou Y, Liu H, Li S, Li Z, Liu Z, Liu C, Pan Y, Cai X, Meng X, Wang Y, Li H, Jiang Y, Wang S, Niu H, Wei T, Wang Y, Liu T. Disrupted structural network resilience in atherosclerosis: A large-scale cohort study. Brain Res 2025; 1859:149653. [PMID: 40252894 DOI: 10.1016/j.brainres.2025.149653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/22/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Atherosclerosis is a major factor in cognitive decline among aging individuals and is frequently linked to the accumulation of white matter hyperintensities. Brain resilience, which represents the brain's capacity to withstand external disruptions, remains poorly understood in terms of how atherosclerosis impacts it and, in turn, influences cognition. Here, we investigated the relationship between atherosclerosis, white matter hyperintensities, and structural network resilience, along with their combined effects on cognitive performance. METHODS We utilized data from the large-scale community cohort Polyvascular Evaluation for Cognitive Impairment and Vascular Events (n = 2160). Whole-brain structural connections were constructed, and structural disconnections were simulated based on white matter hyperintensities. SNR, serving as a marker to quantify structural network resilience, is defined by the similarity of hub nodes between the original network and its disconnected counterpart. RESULTS SNR showed higher odds ratios compared to white matter hyperintensities in relation to arterial status. Additionally, chain mediation analysis indicated that cognitive decline associated with atherosclerosis was partially mediated by both white matter hyperintensities and structural network resilience. Atherosclerosis accelerates the degradation of brain structural network resilience as age increases. CONCLUSIONS These findings suggest that SNR could offer complementary insights into cognitive decline caused by atherosclerosis and serve as a potential biomarker of brain health in atherosclerotic conditions. Additionally, SNR may act as an indicator for guiding the selection of future therapies for atherosclerosis.
Collapse
Affiliation(s)
- Hui Tang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wanlin Zhu
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Jing
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yijun Zhou
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Hao Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shiping Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zixiao Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ziyang Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Chang Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yuesong Pan
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xueli Cai
- Department of Neurology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Xia Meng
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yilong Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yong Jiang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Suying Wang
- Cerebrovascular Research Lab, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Haijun Niu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Tiemin Wei
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Tao Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| |
Collapse
|
2
|
Khowdiary MM, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhenawy AA, Rashwan EK, Alexiou A, Papadakis M, Fetoh MEAE, Batiha GES. The Peripheral Amyloid-β Nexus: Connecting Alzheimer's Disease with Atherosclerosis through Shared Pathophysiological Mechanisms. Neuromolecular Med 2025; 27:20. [PMID: 40032716 PMCID: PMC11876215 DOI: 10.1007/s12017-025-08836-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two chronic diseases with seemingly distinct pathologies. However, emerging research points to a bidirectional relationship driven by common mechanisms, such as inflammation, oxidative stress, and dysregulation of Amyloid-Beta (Aβ). This review focuses on the role of Aβ as a critical molecular link between AD and AS, emphasizing its contribution to neuronal impairment and vascular damage. Specifically, peripheral Aβ produced in the pancreas and skeletal muscle tissues exacerbates AS by promoting endothelial dysfunction and insulin resistance (IR). Furthermore, AS accelerates AD progression by impairing cerebral blood flow and inducing chronic hypoxia, causing Aβ accumulation. This review critically evaluates recent findings, highlighting inconsistencies in clinical studies and suggesting future research directions. Understanding the bidirectional influence of AD and AS could pave the way for novel therapeutic approaches targeting shared molecular pathways, particularly emphasizing Aβ clearance and inflammation.
Collapse
Affiliation(s)
- Manal M Khowdiary
- Department of Chemistry, Faculty of Applied Science, Lieth Collage, Umm Al-Qura University, 24382, Makkah, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO. Box13, Kufa, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
- Chemistry Department, Faculty of Science, AlBaha University, 65731, Al Bahah, Saudi Arabia
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Jouf University, Akaka, Saudi Arabia
| | - Athanasios Alexiou
- Department of Research & Development, Funogen, 11741, Athens, Attiki, Greece
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
| | - Marios Papadakis
- University Hospital, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammed E Abo-El Fetoh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
3
|
Roumeliotis S, Kontogiorgos I, de Vries F, Maresz K, Jeanne JF, Leivaditis K, Schurgers LJ. The role of vitamin K2 in cognitive impairment: linking vascular health to brain health. Front Aging Neurosci 2025; 16:1527535. [PMID: 39881683 PMCID: PMC11775153 DOI: 10.3389/fnagi.2024.1527535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Cognitive impairment, marked by a decline in essential mental aspects such as attention, memory, and problem-solving, is significantly correlated with advancing age. This condition presents a major challenge for the elderly, adversely affecting quality of life, diminishing independence, and imposing substantial burdens on healthcare systems. Recent research indicates that vitamin K2 may be vital for preserving brain health and cognitive function. Traditionally recognized primarily for its role in blood coagulation, vitamin K has emerged in recent years as a nutrient with diverse biological effects essential for healthy aging. A growing body of evidence from both observational and interventional studies underscores the pivotal role of vitamin K2 in mitigating arterial calcification. This mechanism may link vascular health to cognitive function, suggesting that vitamin K2 could play a critical role in the prevention of cognitive impairment in aging populations.
Collapse
Affiliation(s)
- Stefanos Roumeliotis
- Second Department of Nephrology, School of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Kontogiorgos
- Second Department of Nephrology, School of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Femke de Vries
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | | | - Jean-François Jeanne
- Gnosis by Lesaffre, Lesaffre International, R&D Department, Marcq-En-Baroeul, France
| | - Konstantinos Leivaditis
- Second Department of Nephrology, School of Medicine, AHEPA Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Leon J. Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| |
Collapse
|
4
|
Vestergaard MB, Bakhtiari A, Osler M, Mortensen EL, Lindberg U, Law I, Lauritzen M, Benedek K, Larsson HBW. The cerebral blood flow response to neuroactivation is reduced in cognitively normal men with β-amyloid accumulation. Alzheimers Res Ther 2025; 17:4. [PMID: 39754275 PMCID: PMC11699738 DOI: 10.1186/s13195-024-01652-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Accumulation of β-amyloid (Aβ) in the brain is a hallmark of Alzheimer's Disease (AD). Cerebral deposition of Aβ initiates deteriorating pathways which eventually can lead to AD. However, the exact mechanisms are not known. A possible pathway could be that Aβ affects the cerebral vessels, causing inadequate cerebrovascular function. In the present study, we examined if Aβ accumulation is associated with a reduced cerebral blood flow response (CBF) to neuronal activation by visual stimulation (ΔCBFVis.Act.) in cognitively normal subjects from the Metropolit Danish Male Birth Cohort. METHODS 64 subjects participated in the present study. ΔCBFVis.Act. was measured using arterial spin labelling (ASL) combined with blood-oxygen-level-dependent (BOLD) MRI. Neuronal activation was obtained by visual stimulation by a flickering checkerboard presented on a screen in the MRI-scanner. Brain Aβ accumulation and cerebral glucose metabolism were assessed by PET imaging using the radiotracers [11C]Pittsburgh Compound-B (PiB) and [18F]Fluorodeoxyglucose (FDG), respectively. Cortical thickness was measured from structural MRI. RESULTS ΔCBFVis.Act. correlated negatively ( β = -32.1 [95% confidence interval (CI): -60.2; -4.1], r = -0.30, p = 0.025) with PiB standardized uptake value ratio (SUVr) in the brain regions activated by visual stimulation. ΔCBFVis.Act. did not correlate with FDG SUVr ( β = 1.9 [CI: -23.8; 27.6], r = 0.02, p = 0.88) or cortical thickness ( β = 10.3 [CI: -8.4; 29.0], r = 0.15, p = 0.27) in the activated brain regions. Resting CBF did not correlate with PiB SUVr neither in the regions activated by visual stimulation ( β = -17.8 [CI:-71.9; 36.2], r =- 0.09, p = 0.51) nor in the remaining cortex ( β = 5.2 [CI:-3.9; 14.2], r = 0.15, p = 0.26). CONCLUSION We found a correlation between high PiB SUVr and reduced CBF response to neuronal activation, indicating a link between Aβ accumulation and impaired cerebrovascular function. The impairment was not associated with cortical thinning or hypometabolism, suggesting that Aβ accumulation affecting brain vessel function could be a very early pathology leading to neurodegenerative disease.
Collapse
Affiliation(s)
- Mark Bitsch Vestergaard
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark.
| | - Aftab Bakhtiari
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Merete Osler
- Center for Clinical Research and Prevention, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | | | - Ulrich Lindberg
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Martin Lauritzen
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krisztina Benedek
- Department of Clinical Neurophysiology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Henrik Bo Wiberg Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Valdemar Hansens Vej 1-23, Glostrup, 2600, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Takahashi MKN, Paradela RS, Grinberg LT, Leite REP, Farias-Itao DS, Paes VR, Braga ME, Naslavsky MS, Zatz M, Jacob-Filho W, Nitrini R, Pasqualucci CA, Suemoto CK. Hypertension may associate with cerebral small vessel disease and infarcts through the pathway of intracranial atherosclerosis. Neurobiol Aging 2025; 145:84-95. [PMID: 39541803 PMCID: PMC11864294 DOI: 10.1016/j.neurobiolaging.2024.11.001] [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: 04/21/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Hypertension, a major modifiable risk factor for cardiovascular diseases, is linked to late-life neurocognitive disorders such as vascular dementia and Alzheimer's disease (AD). This study explores the associations between hypertension, intracranial atherosclerotic disease (ICAD), cerebral small vessel disease (cSVD), and Alzheimer's disease neuropathologic change (ADNC) in a large community-based autopsy study. This cross-sectional study used data from the Biobank for Aging Studies of the University of São Paulo Medical School. Sociodemographic and clinical information was gathered from a reliable next-of-kin informant. Neurofibrillary tangles, neuritic plaques, lacunar infarcts, hyaline arteriolosclerosis, and cerebral amyloid angiopathy were evaluated. Causal mediation analyses with natural effect models were performed to examine indirect associations of hypertension with cerebrovascular pathologies and ADNC through morphometric measurements of intracranial artery lumen obstruction. Hypertensive participants (n = 354) presented a higher rate of stenosed arteries (obstruction ≥ 50 %), critically stenosed arteries (obstruction ≥ 70 %), and more severe ICAD, shown by higher maximum and mean obstruction indexes compared to nonhypertensive participants (n = 166). These measurements of atherosclerosis were associated with neurofibrillary tangles and cSVD lesions. Hypertension was indirectly associated with hyaline arteriolosclerosis and lacunar infarcts through the pathway of ICAD. Presenting hypertension indirectly increased the odds of displaying hyaline arteriolosclerosis by 26 % (95 % CI: 1.08, 1.45, p = 0.002) and lacunar infarcts by 17 % (95 % CI: 1.01, 1.35, p = 0.029). Cognitive and APOE ε4 carrier status did not alter the investigated associations. In this community sample, hypertension was indirectly associated with cSVD through ICAD.
Collapse
Affiliation(s)
| | - Regina Silva Paradela
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Lea Tenenholz Grinberg
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, SP 01246-903, Brazil; Memory and Aging Center, Weill Institute for Neurosciences, Dept. of Neurology, University of California San Francisco, CA 94158, USA
| | | | | | - Vitor Ribeiro Paes
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, SP 01246-903, Brazil
| | - Maria Eduarda Braga
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Michel Satya Naslavsky
- Human Genome and Stem Cell Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Mayana Zatz
- Human Genome and Stem Cell Center, Biosciences Institute, University of Sao Paulo, Sao Paulo 05508-090, Brazil
| | - Wilson Jacob-Filho
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil
| | - Ricardo Nitrini
- Department of Neurology, University of Sao Paulo Medical School, SP 01246-903, Brazil
| | | | - Claudia Kimie Suemoto
- Division of Geriatrics, University of Sao Paulo Medical School, Sao Paulo 01246-903, Brazil.
| |
Collapse
|
6
|
Barisano G, Iv M, Choupan J, Hayden-Gephart M. Robust, fully-automated assessment of cerebral perivascular spaces and white matter lesions: a multicentre MRI longitudinal study of their evolution and association with risk of dementia and accelerated brain atrophy. EBioMedicine 2025; 111:105523. [PMID: 39721217 PMCID: PMC11732520 DOI: 10.1016/j.ebiom.2024.105523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Perivascular spaces (PVS) on brain MRI are surrogates for small parenchymal blood vessels and their perivascular compartment, and may relate to brain health. However, it is unknown whether PVS can predict dementia risk and brain atrophy trajectories in participants without dementia, as longitudinal studies on PVS are scarce and current methods for PVS assessment lack robustness and inter-scanner reproducibility. METHODS We developed a robust algorithm to automatically assess PVS count and size on clinical MRI, and investigated 1) their relationship with dementia risk and brain atrophy in participants without dementia, 2) their longitudinal evolution, and 3) their potential use as a screening tool in simulated clinical trials. We analysed 46,478 clinical measurements of cognitive functioning and 20,845 brain MRI scans from 10,004 participants (71.1 ± 9.7 years-old, 56.6% women) from three publicly available observational studies on ageing and dementia (the Alzheimer's Disease Neuroimaging Initiative, the National Alzheimer's Coordinating Centre database, and the Open Access Series of Imaging Studies). Clinical and MRI data collected between 2004 and 2022 were analysed with consistent methods, controlling for confounding factors, and combined using mixed-effects models. FINDINGS Our fully-automated method for PVS assessment showed excellent inter-scanner reproducibility (intraclass correlation coefficients >0.8). Fewer PVS and larger PVS diameter at baseline predicted higher dementia risk and accelerated brain atrophy. Longitudinal trajectories of PVS markers differed significantly in participants without dementia who converted to dementia compared with non-converters. In simulated placebo-controlled trials for treatments targeting cognitive decline, screening out participants at low risk of dementia based on our PVS markers enhanced the power of the trial independently of Alzheimer's disease biomarkers. INTERPRETATION These robust cerebrovascular markers predict dementia risk and brain atrophy and may improve risk-stratification of patients, potentially reducing cost and increasing throughput of clinical trials to combat dementia. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
| | - Michael Iv
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Jeiran Choupan
- Laboratory of Neuro Imaging, University of Southern California, Los Angeles, CA, USA; NeuroScope Inc., New York, NY, USA
| | | |
Collapse
|
7
|
Yang D, Cherian L, Arfanakis K, Schneider JA, Aggarwal NT, Gutierrez J. Intracranial atherosclerotic disease and neurodegeneration: a narrative review and plausible mechanisms. J Stroke Cerebrovasc Dis 2024; 33:108015. [PMID: 39303868 PMCID: PMC11570339 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION Intracranial atherosclerotic disease (ICAD) of the large cerebral arteries, a leading cause of stroke worldwide, is increasingly implicated in cognitive impairment and neurodegeneration among the general population; however, the underlying pathophysiologic mechanisms in this relationship remain unknown. METHODS In this narrative review, we aim to provide an overview of the epidemiology and pathophysiology of ICAD, the evidence that relates ICAD to neurodegeneration, putative mechanisms, and future research directions. We synthesized available evidence on PubMed up to August 2024. RESULTS AND CONCLUSIONS ICAD, a common cause of stroke, is characterized as a chronic, inflammatory, fibroproliferative disease of the cerebral large arteries. Numerous lines of evidence have related ICAD to clinical, neuroimaging, and pathology-based markers of cognitive impairment and Alzheimer's disease; however, little data exists on plausible pathophysiological links. Based on ongoing and adjacent work, we hypothesize hypoperfusion, arterial stiffness, and inflammation to play a role, but further research is needed. Conventional classification of ICAD often infers from symptomatic coronary artery disease and relies on degree of luminal stenosis, but unique anatomic features of the intracranial circulation may be relevant and a more comprehensive description that includes arterial wall features and plaque morphology may be needed to fully understand its relationship with cognitive impairment and neurodegeneration.
Collapse
Affiliation(s)
- Dixon Yang
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| | - Laurel Cherian
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, IL, USA; Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Julie A Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Neelum T Aggarwal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jose Gutierrez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Prasad M, Goodman D, Gutta S, Sheikh Z, Cabral HJ, Shunyakova J, Sanjiv N, Curley C, Yarala RR, Tsai L, Siegel NH, Chen X, Poulaki V, Alosco ML, Stein TD, Ness S, Subramanian ML. Associations Between Retinal Vascular Occlusions and Dementia. Healthcare (Basel) 2024; 12:2371. [PMID: 39684995 DOI: 10.3390/healthcare12232371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Retinal vascular occlusions, such as retinal vein occlusion (RVO) and retinal artery occlusion (RAO), are associated with cognitive impairment, including dementia. Our objective was to examine the odds of dementia among patients with retinal vascular occlusion. METHODS This cross-sectional study included 474 patients with retinal vascular occlusion and 948 patients without retinal vascular occlusion (comparison group). Patients in the comparison group were age- and sex-matched to those with vascular occlusion. Logistic regression was used to analyze the odds of all-cause dementia, vascular dementia, and Alzheimer's disease after adjusting for demographic, clinical, and ophthalmic covariates. Main outcome measures included the presence of all-cause dementia, vascular dementia, and Alzheimer's disease. RESULTS Patients with RVO (n = 413) had increased odds for all-cause dementia (odds ratio (OR) = 2.32; 95% confidence interval (CI): 1.44-3.75; p < 0.001) and vascular dementia (OR = 3.29; 95% CI: 1.41-7.68; p = 0.006) relative to the comparison group. Patients with central RVO (n = 192) (OR = 2.32; 95% CI: 1.19-4.54; p = 0.014) or branch RVO (n = 221) (OR = 2.68; 95% CI: 1.30-5.50; p = 0.007) had increased odds for all-cause dementia relative to the comparison group. Patients with RAO (n = 61) did not have increased odds of all-cause dementia (OR = 1.01; 95% CI: 0.32-3.26; p = 0.983), vascular dementia (OR = 1.54; 95% CI: 0.22-10.81; p = 0.663), or Alzheimer's disease (OR = 0.32; 95% CI: 0.05-2.20; p = 0.244). CONCLUSIONS A history of any RVO is associated with increased rates of all-cause dementia and vascular dementia independent of shared cardiovascular risk factors. These associations are not seen with a history of RAO, or between any subtype of vascular occlusions and Alzheimer's disease.
Collapse
Affiliation(s)
- Minali Prasad
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Deniz Goodman
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Sanhit Gutta
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Zahra Sheikh
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Howard J Cabral
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jenny Shunyakova
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Nayan Sanjiv
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Cameron Curley
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Rohun Reddy Yarala
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Lynna Tsai
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Nicole H Siegel
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Xuejing Chen
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Vasiliki Poulaki
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Thor D Stein
- VA Boston Healthcare System, Boston, MA 02130, USA
- Boston University Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Boston University CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- VA Bedford Healthcare System, Bedford, MA 01730, USA
| | - Steven Ness
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| | - Manju L Subramanian
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Ophthalmology, Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
9
|
Lam TG, Ross SK, Ciener B, Xiao H, Flaherty D, Lee AJ, Dugger BN, Reddy H, Teich AF. Pathologic subtyping of Alzheimer's disease brain tissue reveals disease heterogeneity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.14.24315458. [PMID: 39484271 PMCID: PMC11527055 DOI: 10.1101/2024.10.14.24315458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
In recent years, multiple groups have shown that what is currently thought of as "Alzheimer's Disease" (AD) may be usefully viewed as several related disease subtypes. As these efforts have continued, a related issue is how common co-pathologies and ethnicity intersect with AD subtypes. The goal of this study was to use a dataset constituting 153 pathologic variables recorded on 666 AD brain autopsies to better define how co-pathologies and ethnicity relate to established AD subtypes. Pathologic clustering suggests 8 subtypes within this cohort, and further analysis reveals that the previously described continuum from hippocampal predominant to hippocampal sparing is well represented in our data. Small vessel disease is overall highest in a cluster with a low hippocampal/cortical tau ratio, and across all clusters small vessel disease segregates separately from Lewy body disease. Two AD clusters are identified with extensive Lewy bodies outside amygdala (one with a high hippocampal/cortical tau ratio and one with a low ratio), and we find an inverse relationship between cortical tau and Lewy body pathology across these two clusters. Finally, we find that brains from persons of Hispanic descent have significantly more AD pathology in multiple neuroanatomic areas. We find that Hispanic ethnicity is not uniformly distributed across clusters, and this is particularly pronounced in clusters with significant Lewy body pathology, where Hispanic donors are only found in a cluster with a low hippocampal/cortical tau ratio. In summary, our analysis of recorded pathologic data across two decades of banked brains reveals new relationships in the patterns of AD-related proteinopathy, co-pathology, and ethnicity, and highlights the utility of pathologic subtyping to classify AD pathology.
Collapse
Affiliation(s)
- Tiffany G. Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sophie K. Ross
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benjamin Ciener
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Harrison Xiao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Delaney Flaherty
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Brittany N. Dugger
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Hasini Reddy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew F. Teich
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Zhao D, Guallar E, Qiao Y, Knopman DS, Palatino M, Gottesman RF, Mosley TH, Wasserman BA. Intracranial Atherosclerotic Disease and Incident Dementia: The ARIC Study (Atherosclerosis Risk in Communities). Circulation 2024; 150:838-847. [PMID: 39087353 PMCID: PMC11513165 DOI: 10.1161/circulationaha.123.067003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 06/26/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Studies of the neurovascular contribution to dementia have largely focused on cerebral small vessel disease (CSVD), but the role of intracranial atherosclerotic disease (ICAD) remains unknown in the general population. The objective of this study was to determine the risk of incident dementia from ICAD after adjusting for CSVD and cardiovascular risk factors in a US community-based cohort. METHODS We acquired brain magnetic resonance imaging examinations from 2011 through 2013 in 1980 Black and White participants in the ARIC study (Atherosclerosis Risk in Communities), a prospective cohort conducted in 4 US communities. Magnetic resonance imaging examinations included high-resolution vessel wall magnetic resonance imaging and magnetic resonance angiography to identify ICAD. Of these participants, 1590 without dementia, without missing covariates, and with adequate magnetic resonance image quality were followed through 2019 for incident dementia. Associations between ICAD and incident dementia were assessed using Cox proportional hazard ratios adjusted for CSVD (characterized by white matter hyperintensities, lacunar infarctions, and microhemorrhages), APOE4 genotype (apolipoprotein E gene ε4), and cardiovascular risk factors. RESULTS The mean age (SD) of study participants was 77.4 (5.2) years. ICAD was detected in 34.6% of participants. After a median follow-up of 5.6 years, 286 participants developed dementia. Compared with participants without ICAD, the fully adjusted hazard ratios (95% CIs) for incident dementia in participants with any ICAD, with ICAD only causing stenosis ≤50%, and with ICAD causing stenosis >50% in ≥1 vessel were 1.57 (1.17-2.11), 1.41 (1.02-1.95), and 1.94 (1.32-2.84), respectively. ICAD was associated with dementia even among participants with low white matter hyperintensities burden, a marker of CSVD. CONCLUSIONS ICAD was associated with an increased risk of incident dementia, independent of CSVD, APOE4 genotype, and cardiovascular risk factors. The increased risk of dementia was evident even among participants with low CSVD burden, a group less likely to be affected by vascular dementia, and in participants with ICAD causing only low-grade stenosis. Our results suggest that ICAD may partially mediate the effect that cardiovascular risk factors have on the brain leading to dementia. Both ICAD and CSVD must be considered to understand the vascular contributions to cognitive decline.
Collapse
Affiliation(s)
- Di Zhao
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Eliseo Guallar
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Ye Qiao
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Maylin Palatino
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Rebecca F. Gottesman
- Stroke Branch, Intramural Research Program, National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Thomas H. Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Bruce A. Wasserman
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
11
|
Stern A, H Frishman W. Connections Between Hypertension, Atherosclerosis, Acute Myocardial Infarction, and Risk of Dementia. Cardiol Rev 2024:00045415-990000000-00300. [PMID: 40359316 DOI: 10.1097/crd.0000000000000739] [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: 05/15/2025]
Abstract
As the percentage of geriatric patients continues to increase in both the United States and globally, the prevalence of both cardiovascular disease and dementia continues to climb. Both dementia and cardiovascular disease are devastating diseases that impose a significant burden economically, socially, and medically on both a local and systemic level. The most common fatal manifestation of cardiovascular disease is acute myocardial infarction, responsible for death in more than 80% of patients with cardiovascular disease. Prominent risk factors for acute myocardial infarction including hypertension and atherosclerosis have been independently associated with an increased risk for cognitive decline and all-cause dementia and Alzheimer disease, separate from vascular dementia. Acute myocardial infarction itself has also been independently associated with an increased incidence of all-cause dementia and Alzheimer disease. It is based on the connection between acute myocardial infarction, its major risk factors, and the incidence of dementia that it is of importance to define and explore the potential role that therapies for these conditions, as well as acute myocardial infarction itself, may play in mitigating the risk of dementia onset and severity. In this review, we assess current therapeutics that exist for atherosclerosis, hypertension and acute myocardial infarction that have been demonstrated to reduce later risk of dementia, and explore the mechanism that underlies the association between the incidence of acute myocardial infarction and the risk of dementia.
Collapse
Affiliation(s)
- Avi Stern
- From the Department of Medicine, New York Medical College, Valhalla, NY
| | | |
Collapse
|
12
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
14
|
Barisano G, Iv M, on behalf of the Alzheimer’s Disease Neuroimaging Initiative, Choupan J, Hayden-Gephart M. Cerebral perivascular spaces as predictors of dementia risk and accelerated brain atrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.25.24306324. [PMID: 38712073 PMCID: PMC11071547 DOI: 10.1101/2024.04.25.24306324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Cerebral small vessel disease, an important risk factor for dementia, lacks robust, in vivo measurement methods. Perivascular spaces (PVS) on brain MRI are surrogates for small parenchymal blood vessels and their perivascular compartment, and may relate to brain health. We developed a novel, robust algorithm to automatically assess PVS count and size on MRI, and investigated their relationship with dementia risk and brain atrophy. We analyzed 46,478 clinical measurements of cognitive functioning and 20,845 brain MRI scans from 10,004 participants (71.1±9.7 years-old, 56.6% women). Fewer PVS and larger PVS diameter at baseline were associated with higher dementia risk and accelerated brain atrophy. Longitudinal trajectories of PVS markers were significantly different in non-demented individuals who converted to dementia compared with non-converters. In simulated placebo-controlled trials for treatments targeting cognitive decline, screening out participants less likely to develop dementia based on our PVS markers enhanced the power of the trial. These novel radiographic cerebrovascular markers may improve risk-stratification of individuals, potentially reducing cost and increasing throughput of clinical trials to combat dementia.
Collapse
Affiliation(s)
| | - Michael Iv
- Department of Radiology, Stanford University, Stanford, CA, USA
| | | | - Jeiran Choupan
- Laboratory of Neuro Imaging, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
15
|
Yang Y, Zhu D, Qi R, Chen Y, Sheng B, Zhang X. Association between Intake of Edible Mushrooms and Algae and the Risk of Cognitive Impairment in Chinese Older Adults. Nutrients 2024; 16:637. [PMID: 38474765 DOI: 10.3390/nu16050637] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Previous studies have investigated the association between diet and cognitive impairment, yet there is limited investigation into the link between edible mushrooms and algae intake and cognitive decline. This study aims to explore the association between edible mushrooms and algae intake and the risk of cognitive impairment in individuals aged 65 years and above in China. Cross-sectional data from the 2018 Chinese Longitudinal Healthy Longevity Survey (CLHLS) formed the basis of this study. Edible mushrooms and algae intake was evaluated using a simplified food frequency questionnaire (FFQ) and cognitive function was assessed using the Mini-Mental State Examination (MMSE). A binary logistic regression model was used to evaluate odds ratios (ORs) and 95% confidence intervals (CIs), with subgroup analysis conducted. Among 14,150 older adults, the average age was (85.33 ± 11.55), with a cognitive impairment prevalence of 22.7; multi-model adjustments showed a 25.3% lower probability of cognitive impairment for those occasionally consuming edible mushrooms and algae (OR: 0.747, 95% CI: 0.675~0.826). Furthermore, a 29% lower risk was observed in those with daily intake (OR: 0.710, 95% CI: 0.511~0.987). Subgroup analysis demonstrated significant risk reduction in women (OR: 0.589, 95% CI: 0.375~0.925, p = 0.022), individuals with disability in activities of daily living (OR: 0.568, 95% CI: 0.367~0.878, p = 0.011), and those with low social activity levels (OR: 0.671, 95% CI: 0.473~0.950, p = 0.025). This study concludes that edible mushrooms and algae intake significantly impacts the risk of cognitive impairment in older adults. These results provide insights and impetus for further research into this area. Additional cohort studies or intervention trials are necessary to confirm the potential benefits of edible mushrooms and algae in promoting cognitive health.
Collapse
Affiliation(s)
- Yun Yang
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Danni Zhu
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Ran Qi
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Yanchun Chen
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Baihe Sheng
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
| | - Xinyu Zhang
- School of Public Health, Tianjin Medical University, Tianjin 300070, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
16
|
König M, Palmer K, Malsch C, Steinhagen-Thiessen E, Demuth I. Polyvascular atherosclerosis and renal dysfunction increase the odds of cognitive impairment in vascular disease: findings of the LipidCardio study. Eur J Med Res 2024; 29:141. [PMID: 38388510 PMCID: PMC10882759 DOI: 10.1186/s40001-024-01734-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
INTRODUCTION Growing evidence suggests a causal role for atherosclerotic vascular disease in cognitive impairment and dementia. Atherosclerosis may present as monovascular disease (monoVD) or as widespread polyvascular atherosclerotic disease (polyVD). Evidence on the relationship between monoVD or polyVD and cognitive impairment is limited. METHODS We conducted a cross-sectional analysis of baseline data from the LipidCardio Study. The main outcome measure was the presence of cognitive impairment, defined as a Mini-Mental State Examination (MMSE) score < 26. RESULTS The mean age was 71.5 years, 30.3% were female, 17.3% had no evidence of large-vessel atherosclerosis, 71.1% had monoVD, and 11.7% had polyVD, defined as the presence of atherosclerosis in ≥ 2 vascular territories (coronary, cerebral, aortic, or lower extremity). A total of 21.6% had cognitive impairment according to the prespecified cutoff (MMSE < 26). Overall, the odds of cognitive impairment increased for each additional vascular territory affected by atherosclerosis [adjusted odds ratio 1.76, 95% confidence interval (CI) 1.21-2.57, p = 0.003]. Furthermore, there was evidence for an interaction between vascular disease and chronic kidney disease (CKD). The odds of cognitive impairment were not greater in the monoVD subgroup compared to those without any atherosclerosis, if CKD was absent (OR 0.98, 95% CI 0.48-2.10; p = 0.095), while the odds ratio (OR) of cognitive impairment with polyVD compared to no atherosclerosis was 2.71 (95% CI 1.10-6.92; p = 0.031). In contrast, in patients with CKD, both monoVD and polyVD were associated with significantly higher odds of cognitive impairment than no atherosclerosis. CONCLUSIONS PolyVD is associated with increased odds of cognitive impairment. MonoVD is associated with cognitive impairment only in the presence of CKD.
Collapse
Affiliation(s)
- Maximilian König
- Department of Internal Medicine D-Geriatrics, Universitätsmedizin Greifswald, Walther-Rathenau-Str. 49, 17475, Greifswald, Mecklenburg-Vorpommern, Germany.
| | - Katie Palmer
- Department of Clinical Geriatrics, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Carolin Malsch
- Institute for Mathematics and Computer Science, University of Greifswald, Greifswald, Germany
| | - Elisabeth Steinhagen-Thiessen
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- BCRT-Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
17
|
Samy MVG, Perumal S. Systems pharmacology and multi-scale mechanism of Enicostema axillare bioactives in treating Alzheimer disease. Inflammopharmacology 2024; 32:575-593. [PMID: 37845599 DOI: 10.1007/s10787-023-01348-0] [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: 08/15/2023] [Accepted: 09/19/2023] [Indexed: 10/18/2023]
Abstract
As a progressive neurological disease with increased morbidity and mortality, Alzheimer Disease (AD) is characterized by neuron damage that controls memory and mental functions. Enicostema axillare (EA), an herb with a history of combativeness and effectiveness in treating Rheumatoid Arthritis, Cancer, and Diabetes, is used in Indian folk medicine from a holistic point of view. Though the herb is used for many illnesses, the molecular mechanism of its bioactive on AD has not been deciphered by intricate research. A unique pharmacology approach based on ADME drug screening and targeting, pathway enrichment (GO and KEGG), and network pharmacology, was established to explore the molecular mechanisms of E. axillare (EA) bioactive compounds for the treatment of AD. In brief, we bring to light the three active compounds of EA and seven potential molecular targets of AD, which are mainly implicated in four signaling pathways, i.e., MAPK, Apoptosis, neurodegeneration, and the TNF pathway. Moreover, the network analysis of the active compounds, molecular targets, and their pathways reveals the pharmacological nature of the compounds. Further, molecular docking studies were carried out to explore the interactions between the EA bioactive compounds and the targets and examine the binding affinity. The outcome of the work reflects the potential therapeutic effects of the compounds for treating AD through the modulation of the key proteins, which further corroborates the reliability of our network pharmacology analysis. This study not only helps in understanding the molecular mechanism of the drugs but also helps in finding and sorting new drugs for the treatment of AD, and other complex diseases through modern medicine.
Collapse
Affiliation(s)
| | - Sasidharan Perumal
- Cell and Molecular Biology Division, Biome Live Analytical Center, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
18
|
Sabayan B, Goudarzi R, Ji Y, Borhani‐Haghighi A, Olson‐Bullis BA, Murray AM, Sedaghat S. Intracranial Atherosclerosis Disease Associated With Cognitive Impairment and Dementia: Systematic Review and Meta-Analysis. J Am Heart Assoc 2023; 12:e032506. [PMID: 37955546 PMCID: PMC10727275 DOI: 10.1161/jaha.123.032506] [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: 09/03/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Intracranial atherosclerosis disease (ICAD) alters cerebrovascular hemodynamics and brain structural integrity. Multiple studies have evaluated the link between ICAD and cognitive impairment, with mixed results. This study aims to systematically review and summarize the current evidence on this link. METHODS AND RESULTS PubMed, EMBASE, PsycInfo, and Web of Science were searched from 2000 to 2023 without language restriction. Cross-sectional and prospective cohort studies as well as postmortem studies were included. Studies containing data on the link between ICAD, defined as at least 50% stenosis in 1 intracranial vessel, and cognitive impairment and dementia were screened by 2 independent reviewers. A total of 22 (17 observational and 5 postmortem) unique studies, comprising 11 184 individuals (average age range, 59.8-87.6 years; 45.7% women; 36.5% Asian race), were included in the systematic review. Seven of 10 cross-sectional studies and 5 of 7 prospective studies showed a significant association between ICAD and cognitive impairment. In the pooled analysis, ICAD was associated with greater cognitive impairment (measure of association, 1.87 [95% CI, 1.49-2.35]). Meta-regression analyses did not show a significant impact of age, sex, and race. All postmortem studies showed that patients with Alzheimer disease and vascular dementia had a higher burden of ICAD compared with controls. CONCLUSIONS This study shows that ICAD is associated with cognitive impairment and dementia across age, sex, and race groups. Our findings may underscore the need to develop individualized dementia preventive care plans in patients with ICAD.
Collapse
Affiliation(s)
- Behnam Sabayan
- Department of Neurology, Hennepin Healthcare Research InstituteHennepin County Medical CenterMinneapolisMN
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Roham Goudarzi
- Faculty of ScienceUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Yuekai Ji
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| | | | | | - Anne M. Murray
- Berman Center for Outcomes and Clinical Research and Geriatrics Division, Department of MedicineHennepin Healthcare Research InstituteMinneapolisMN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| |
Collapse
|
19
|
Alkhalifa AE, Al-Ghraiybah NF, Odum J, Shunnarah JG, Austin N, Kaddoumi A. Blood-Brain Barrier Breakdown in Alzheimer's Disease: Mechanisms and Targeted Strategies. Int J Mol Sci 2023; 24:16288. [PMID: 38003477 PMCID: PMC10671257 DOI: 10.3390/ijms242216288] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
The blood-brain barrier (BBB) is a unique and selective feature of the central nervous system's vasculature. BBB dysfunction has been observed as an early sign of Alzheimer's Disease (AD) before the onset of dementia or neurodegeneration. The intricate relationship between the BBB and the pathogenesis of AD, especially in the context of neurovascular coupling and the overlap of pathophysiology in neurodegenerative and cerebrovascular diseases, underscores the urgency to understand the BBB's role more deeply. Preserving or restoring the BBB function emerges as a potentially promising strategy for mitigating the progression and severity of AD. Molecular and genetic changes, such as the isoform ε4 of apolipoprotein E (ApoEε4), a significant genetic risk factor and a promoter of the BBB dysfunction, have been shown to mediate the BBB disruption. Additionally, receptors and transporters like the low-density lipoprotein receptor-related protein 1 (LRP1), P-glycoprotein (P-gp), and the receptor for advanced glycation end products (RAGEs) have been implicated in AD's pathogenesis. In this comprehensive review, we endeavor to shed light on the intricate pathogenic and therapeutic connections between AD and the BBB. We also delve into the latest developments and pioneering strategies targeting the BBB for therapeutic interventions, addressing its potential as a barrier and a carrier. By providing an integrative perspective, we anticipate paving the way for future research and treatments focused on exploiting the BBB's role in AD pathogenesis and therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Amal Kaddoumi
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S. Donahue Dr., Auburn, AL 36849, USA; (A.E.A.); (N.F.A.-G.); (J.O.); (J.G.S.); (N.A.)
| |
Collapse
|
20
|
Davidson CG, Woodford SJ, Mathur S, Valle DB, Foster D, Kioutchoukova I, Mahmood A, Lucke-Wold B. Investigation into the vascular contributors to dementia and the associated treatments. EXPLORATION OF NEUROSCIENCE 2023; 2:224-237. [PMID: 37981945 PMCID: PMC10655228 DOI: 10.37349/en.2023.00023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/20/2023] [Indexed: 11/21/2023]
Abstract
As the average lifespan has increased, memory disorders have become a more pressing public health concern. However, dementia in the elderly population is often neglected in light of other health priorities. Therefore, expanding the knowledge surrounding the pathology of dementia will allow more informed decision-making regarding treatment within elderly and older adult populations. An important emerging avenue in dementia research is understanding the vascular contributors to dementia. This review summarizes potential causes of vascular cognitive impairment like stroke, microinfarction, hypertension, atherosclerosis, blood-brain-barrier dysfunction, and cerebral amyloid angiopathy. Also, this review address treatments that target these vascular impairments that also show promising results in reducing patient's risk for and experience of dementia.
Collapse
Affiliation(s)
| | | | - Shreya Mathur
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Devon Foster
- University of Central Florida, Orlando, FL 32816, USA
| | | | - Arman Mahmood
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
21
|
Anwar MM, Mabrouk AA. Hepatic and cardiac implications of increased toxic amyloid-beta serum level in lipopolysaccharide-induced neuroinflammation in rats: new insights into alleviating therapeutic interventions. Inflammopharmacology 2023; 31:1257-1277. [PMID: 37017850 DOI: 10.1007/s10787-023-01202-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 04/06/2023]
Abstract
Neuroinflammation is a devastating predisposing factor for Alzheimer's disease (AD). A number of clinical findings have reported peripheral disorders among AD patients. Amyloid beta (Aβ) is a toxic physiological aggregate that serves as a triggering factor for hepatic and cardiac disorders related to neurotoxicity. As a drawback of Aβ excessive accumulation in the brain, part of Aβ is believed to readily cross the blood-brain barrier (BBB) into the peripheral circulation resulting in serious inflammatory and toxic cascades acting as a direct bridge to cardiac and hepatic pathophysiology. The main aim is to find out whether neuroinflammation-related AD may result in cardiac and liver dysfunctions. Potential therapeutic interventions are also suggested to alleviate AD's cardiac and hepatic defects. Male rats were divided into: control group I, lipopolysaccharide (LPS)-neuroinflammatory-induced group II, LPS-neuroinflammatory-induced group treated with sodium hydrogen sulphide donor (NaHS) (group III), and LPS-neuroinflammatory-induced group treated with mesenchymal stem cells (MSCs) (group IV). Behavior and histopathological studies were conducted in addition to the estimation of different biological biomarkers. It was revealed that the increased toxic Aβ level in blood resulted in cardiac and hepatic malfunctions as a drawback of exaggerated inflammatory cascades. The administration of NaHS and MSCs proved their efficiency in combating neuroinflammatory drawbacks by hindering cardiac and hepatic dysfunctions. The consistent direct association of decreased heart and liver functions with increased Aβ levels highlights the direct involvement of AD in other organ complications. Thereby, these findings will open new avenues for combating neuroinflammatory-related AD and long-term asymptomatic toxicity.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt.
| | - Abeer A Mabrouk
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| |
Collapse
|
22
|
Nichols E, Merrick R, Hay SI, Himali D, Himali JJ, Hunter S, Keage HAD, Latimer CS, Scott MR, Steinmetz JD, Walker JM, Wharton SB, Wiedner CD, Crane PK, Keene CD, Launer LJ, Matthews FE, Schneider J, Seshadri S, White L, Brayne C, Vos T. The prevalence, correlation, and co-occurrence of neuropathology in old age: harmonisation of 12 measures across six community-based autopsy studies of dementia. THE LANCET. HEALTHY LONGEVITY 2023; 4:e115-e125. [PMID: 36870337 PMCID: PMC9977689 DOI: 10.1016/s2666-7568(23)00019-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Population-based autopsy studies provide valuable insights into the causes of dementia but are limited by sample size and restriction to specific populations. Harmonisation across studies increases statistical power and allows meaningful comparisons between studies. We aimed to harmonise neuropathology measures across studies and assess the prevalence, correlation, and co-occurrence of neuropathologies in the ageing population. METHODS We combined data from six community-based autopsy cohorts in the US and the UK in a coordinated cross-sectional analysis. Among all decedents aged 80 years or older, we assessed 12 neuropathologies known to be associated with dementia: arteriolosclerosis, atherosclerosis, macroinfarcts, microinfarcts, lacunes, cerebral amyloid angiopathy, Braak neurofibrillary tangle stage, Consortium to Establish a Registry for Alzheimer's disease (CERAD) diffuse plaque score, CERAD neuritic plaque score, hippocampal sclerosis, limbic-predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC), and Lewy body pathology. We divided measures into three groups describing level of confidence (low, moderate, and high) in harmonisation. We described the prevalence, correlations, and co-occurrence of neuropathologies. FINDINGS The cohorts included 4354 decedents aged 80 years or older with autopsy data. All cohorts included more women than men, with the exception of one study that only included men, and all cohorts included decedents at older ages (range of mean age at death across cohorts 88·0-91·6 years). Measures of Alzheimer's disease neuropathological change, Braak stage and CERAD scores, were in the high confidence category, whereas measures of vascular neuropathologies were in the low (arterioloscerosis, atherosclerosis, cerebral amyloid angiopathy, and lacunes) or moderate (macroinfarcts and microinfarcts) categories. Neuropathology prevalence and co-occurrence was high (2443 [91%] of 2695 participants had more than one of six key neuropathologies and 1106 [41%] of 2695 had three or more). Co-occurrence was strongly but not deterministically associated with dementia status. Vascular and Alzheimer's disease features clustered separately in correlation analyses, and LATE-NC had moderate associations with Alzheimer's disease measures (eg, Braak stage ρ=0·31 [95% CI 0·20-0·42]). INTERPRETATION Higher variability and more inconsistency in the measurement of vascular neuropathologies compared with the measurement of Alzheimer's disease neuropathological change suggests the development of new frameworks for the measurement of vascular neuropathologies might be helpful. Results highlight the complexity and multi-morbidity of the brain pathologies that underlie dementia in older adults and suggest that prevention efforts and treatments should be multifaceted. FUNDING Gates Ventures.
Collapse
Affiliation(s)
- Emma Nichols
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA.
| | - Richard Merrick
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Simon I Hay
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | | | - Jayandra J Himali
- Framingham Heart Study, Framingham, MA, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sally Hunter
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Hannah A D Keage
- Cognitive Ageing and Impairment Neurosciences Lab, Justice and Society, University of South Australia, Adelaide, SA, Australia
| | - Caitlin S Latimer
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Matthew R Scott
- Framingham Heart Study, Framingham, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jaimie D Steinmetz
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Jamie M Walker
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mt Sinai, New York, NY, USA
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Crystal D Wiedner
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Fiona E Matthews
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Julie Schneider
- Rush Alzheimer's Disease Center, Chicago, IL, USA; Rush University Medical Center, Chicago, IL, USA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, MA, USA; Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA; Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Lon White
- Pacific Health Research and Education Institute, Honolulu, HI, USA
| | - Carol Brayne
- Cambridge Public Health, University of Cambridge, Cambridge, UK
| | - Theo Vos
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Bickel MA, Csik B, Gulej R, Ungvari A, Nyul-Toth A, Conley SM. Cell non-autonomous regulation of cerebrovascular aging processes by the somatotropic axis. Front Endocrinol (Lausanne) 2023; 14:1087053. [PMID: 36755922 PMCID: PMC9900125 DOI: 10.3389/fendo.2023.1087053] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023] Open
Abstract
Age-related cerebrovascular pathologies, ranging from cerebromicrovascular functional and structural alterations to large vessel atherosclerosis, promote the genesis of vascular cognitive impairment and dementia (VCID) and exacerbate Alzheimer's disease. Recent advances in geroscience, including results from studies on heterochronic parabiosis models, reinforce the hypothesis that cell non-autonomous mechanisms play a key role in regulating cerebrovascular aging processes. Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) exert multifaceted vasoprotective effects and production of both hormones is significantly reduced in aging. This brief overview focuses on the role of age-related GH/IGF-1 deficiency in the development of cerebrovascular pathologies and VCID. It explores the mechanistic links among alterations in the somatotropic axis, specific macrovascular and microvascular pathologies (including capillary rarefaction, microhemorrhages, impaired endothelial regulation of cerebral blood flow, disruption of the blood brain barrier, decreased neurovascular coupling, and atherogenesis) and cognitive impairment. Improved understanding of cell non-autonomous mechanisms of vascular aging is crucial to identify targets for intervention to promote cerebrovascular and brain health in older adults.
Collapse
Affiliation(s)
- Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anna Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Department of Public Health, Semmelweis University, Budapest, Hungary
- Institute of Biophysics, Biological Research Centre, Eötvös Lorand Research Network (ELKH), Szeged, Hungary
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
24
|
Chen H, Zhou Y, Huang L, Xu X, Yuan C. Multimorbidity burden and developmental trajectory in relation to later‐life dementia: A prospective study. Alzheimers Dement 2022; 19:2024-2033. [PMID: 36427050 DOI: 10.1002/alz.12840] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION This study assessed the associations of multimorbidity burden and its developmental trajectory with later-life dementia. METHODS Among 5923 Health and Retirement Study participants, major chronic conditions including hypertension, diabetes mellitus, cancer, lung diseases, heart disease, stroke, psychological disorders, and arthritis were self- or proxy-reported in 1994-2008. Dementia diagnosis was self- or proxy-reported in 2008-2018. We used Cox regression to assess the associations of multimorbidity with incident dementia. RESULTS During follow-up (median = 8 years), 701 participants developed dementia. Each additional chronic condition in 2008 was related to 15% (confidence interval: 9% to 22%) higher hazard of dementia. Multimorbidity trajectories in 1994-2008 were classified as "rapid growth", "steady growth", "slow growth", and "no new condition" by the group-based trajectory modelling methods. Compared to "no new condition", the "rapid growth" trajectory was related to 32% (3% to 69%) higher dementia risk. CONCLUSIONS Both multimorbidity burden and its developmental trajectory were prospectively associated with risk of dementia.
Collapse
Affiliation(s)
- Hui Chen
- School of Public Health and the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang China
| | - Yaguan Zhou
- School of Public Health and the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang China
| | - Liyan Huang
- School of Public Health and the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang China
| | - Xiaolin Xu
- School of Public Health and the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang China
- School of Public Health Faculty of Medicine The University of Queensland Brisbane Australia
| | - Changzheng Yuan
- School of Public Health and the Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang China
- Department of Nutrition Harvard T.H. Chan School of Public Health Boston Massachusetts USA
| |
Collapse
|
25
|
Zhong T, Qi Y, Li R, Zhou H, Ran B, Wang J, Cai Z. Contribution of intracranial artery stenosis to white matter hyperintensities progression in elderly Chinese patients: A 3-year retrospective longitudinal study. Front Neurol 2022; 13:922320. [PMID: 36212654 PMCID: PMC9539973 DOI: 10.3389/fneur.2022.922320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purposeThere have been controversial results in previous studies for the association between intracranial artery stenosis (ICAS) and white matter hyperintensities (WMHs), and the correlation of ICAS with the progression of WMHs is uncertain. The aim of this study was to investigate the association between ICAS and the progression of WMHs.MethodsIn this retrospective longitudinal study, we enrolled 302 patients aged 60 years and older who had received two brain MRI scans with a 3-year interval and was examined by CTA in the first MRI scan. We measured the stenosis of major intracranial arteries by CTA and assessed the progression of WMHs using the modified Rotterdam Progression scale (mRPS). We performed binary logistic regression analyses and established linear regression model to determine the relationship between the degree of ICAS and the progression of WMHs.ResultsA total of 302 patients were enrolled, of which 48.3% experienced WMHs progression. After adjustment for confounding factors, the patients with Grade 2 ICAS had an OR of 2.8 (95% CI 1.4–5.5), and those with Grade 3 ICAS had an OR of 3.0 (95% CI 1.2–7.3) for the progression of WMHs. The ICAS degree remained associated with PVWMHs but had an attenuated relation to SCWMHs. ICAS severity was significantly associated with WMHs progression scores, higher for Grade 3 ICAS [β (SE) = 0.18 (0.18)] followed by Grade 2 ICAS [β (SE) = 0.10 (0.15)] compared with Grade 1 ICAS.ConclusionsPatients with more severe ICAS are more likely to have WMHs progression and have distinct relevancy to PVWMHs and SCWMHs, which may provide clues for understanding mechanisms of WMHs progression.
Collapse
Affiliation(s)
- Tingting Zhong
- Chongqing Medical University, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Yunwen Qi
- Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Rui Li
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huadong Zhou
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Boli Ran
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
| | - Jiao Wang
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
| | - ZhiYou Cai
- Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
- *Correspondence: ZhiYou Cai
| |
Collapse
|
26
|
Porter T, Sim M, Prince RL, Schousboe JT, Bondonno C, Lim WH, Zhu K, Kiel DP, Hodgson JM, Laws SM, Lewis JR. Abdominal aortic calcification on lateral spine images captured during bone density testing and late-life dementia risk in older women: A prospective cohort study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 26:100502. [PMID: 36213133 PMCID: PMC9535408 DOI: 10.1016/j.lanwpc.2022.100502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
BACKGROUND Dementia after the age of 80 years (late-life) is increasingly common due to vascular and non-vascular risk factors. Identifying individuals at higher risk of late-life dementia remains a global priority. METHODS In prospective study of 958 ambulant community-dwelling older women (≥70 years), lateral spine images (LSI) captured in 1998 (baseline) from a bone density machine were used to assess abdominal aortic calcification (AAC). AAC was classified into established categories (low, moderate and extensive). Cardiovascular risk factors and apolipoprotein E (APOE) genotyping were evaluated. Incident 14.5-year late-life dementia was identified from linked hospital and mortality records. FINDINGS At baseline women were 75.0 ± 2.6 years, 44.7% had low AAC, 36.4% had moderate AAC and 18.9% had extensive AAC. Over 14.5- years, 150 (15.7%) women had a late-life dementia hospitalisation (n = 132) and/or death (n = 58). Compared to those with low AAC, women with moderate and extensive AAC were more likely to suffer late-life dementia hospitalisations (9.3%, 15.5%, 18.3%, respectively) and deaths (2.8%, 8.3%, 9.4%, respectively). After adjustment for cardiovascular risk factors and APOE, women with moderate and extensive AAC had twice the relative hazards of late-life dementia (moderate, aHR 2.03 95%CI 1.38-2.97; extensive, aHR 2.10 95%CI 1.33-3.32), compared to women with low AAC. INTERPRETATION In community-dwelling older women, those with more advanced AAC had higher risk of late-life dementia, independent of cardiovascular risk factors and APOE genotype. Given the widespread use of bone density testing, simultaneously capturing AAC information may be a novel, non-invasive, scalable approach to identify older women at risk of late-life dementia. FUNDING Kidney Health Australia, Healthway Health Promotion Foundation of Western Australia, Sir Charles Gairdner Hospital Research Advisory Committee Grant, National Health and Medical Research Council of Australia.
Collapse
Key Words
- AAC, abdominal aortic calcification
- AAC24, abdominal aortic calcification 24 scale scores
- AD, Alzheimer's disease
- APOE, apolipoprotein E
- ASVD, atherosclerotic vascular disease
- AUC, area under the curve
- Aging
- CAC, coronary artery calcification
- CVD, cardiovascular disease
- DXA, dual-energy X-ray absorptiometry
- Dementia
- Epidemiology
- FRS, Framingham General Cardiovascular Risk Scores
- IDI, integrated discrimination improvement
- Imaging
- LSI, lateral spine imaging
- NRI, net reclassification improvement
- ROC, receiver operator characteristics
- Vascular disease
Collapse
Affiliation(s)
- Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Marc Sim
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - Richard L. Prince
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - John T. Schousboe
- Park Nicollet Clinic and HealthPartners Institute, HealthPartners, Minneapolis, MN, USA
- Division of Health Policy and Management, University of Minnesota, Minneapolis, MN, USA
| | - Catherine Bondonno
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - Wai H. Lim
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Renal Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Kun Zhu
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Douglas P. Kiel
- Marcus Institute for Aging Research, Hebrew SeniorLife, Department of Medicine Beth, Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan M. Hodgson
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Joshua R. Lewis
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
- Centre for Kidney Research, Children's Hospital at Westmead, School of Public Health, Sydney Medical School, the University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
27
|
Park HY, Suh CH, Shim WH, Heo H, Kim WS, Lim JS, Lee JH, Kim HS, Kim SJ. Diagnostic yield of TOF-MRA for detecting incidental vascular lesions in patients with cognitive impairment: An observational cohort study. Front Neurol 2022; 13:958037. [PMID: 36090850 PMCID: PMC9453548 DOI: 10.3389/fneur.2022.958037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Objectives The role of three-dimensional (3D) TOF-MRA in patients with cognitive impairment is not well established. We evaluated the diagnostic yield of 3D TOF-MRA for detecting incidental extra- or intracranial artery stenosis and intracranial aneurysm in this patient group. Methods This retrospective study included patients with cognitive impairment undergoing our brain MRI protocol from January 2013 to February 2020. The diagnostic yield of TOF-MRA for detecting incidental vascular lesions was calculated. Patients with positive TOF-MRA results were reviewed to find whether additional treatment was performed. Logistic regression analysis was conducted to identify the clinical risk factors for positive TOF-MRA findings. Results In total, 1,753 patients (mean age, 70.2 ± 10.6 years; 1,044 women) were included; 199 intracranial aneurysms were detected among 162 patients (9.2%, 162/1,753). A 3D TOF-MRA revealed significant artery stenoses (>50% stenosis) in 162 patients (9.2%, 162/1,753). The overall diagnostic yield of TOF-MRA was 16.8% (294/1,753). Among them, 92 patients (31.3%, 92/294) underwent either medical therapy, endovascular intervention, or surgery. In total, eighty-one patients with stenosis were prescribed with either antiplatelet medications or lipid-lowering agent. In total, fifteen patients (aneurysm: 11 patients, stenosis: 4 patients) were further treated with endovascular intervention or surgery. Thus, the “number needed to scan” was 19 for identifying one patient requiring treatment. Multivariate logistic regression analysis showed that being female (odds ratio [OR] 2.05) and old age (OR 1.04) were the independent risk factors for intracranial aneurysm; being male (OR 1.52), old age (OR 1.06), hypertension (OR 1.78), and ischemic heart disease history (OR 2.65) were the independent risk factors for significant artery stenosis. Conclusions Our study demonstrated the potential benefit of 3D TOF-MRA, given that it showed high diagnostic yield for detecting vascular lesions in patients with cognitive impairment and the considerable number of these lesions required further treatment. A 3D TOF-MRA may be included in the routine MR protocol for the work-up of this patient population, especially in older patients and patients with vascular risk factors.
Collapse
Affiliation(s)
- Ho Young Park
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chong Hyun Suh
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
- *Correspondence: Chong Hyun Suh
| | - Woo Hyun Shim
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hwon Heo
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Woo Seok Kim
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Sung Lim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ho Sung Kim
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang Joon Kim
- Department of Radiology and Research, Asan Medical Center, Institute of Radiology, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Stahr N, Galkina EV. Immune Response at the Crossroads of Atherosclerosis and Alzheimer's Disease. Front Cardiovasc Med 2022; 9:870144. [PMID: 35872901 PMCID: PMC9298512 DOI: 10.3389/fcvm.2022.870144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) and cardiovascular disease (CVD) are pathologies that are characterized by common signatures of vascular dysfunction and chronic inflammation that are accelerated with aging. Importantly, epidemiological studies report an independent interaction between AD and CVD and data suggest that chronic inflammation in CVD may accelerate AD development. Atherosclerosis affects most large to medium sized arteries including those supplying the cerebral circulation. Vascular dysfunction caused by atherosclerosis results in blood brain barrier breakdown, inflammation, an impaired clearance of amyloid-beta (Aβ), and finally ends with neurovascular dysfunction. Numerous data indicate that innate and adaptive immune responses shape atherogenesis and increasing evidence suggests an implication of the immune response in AD progression. Currently, mechanisms by which these two diseases are interconnected with each other are not well-defined. In this review, we discuss the recent advances in our understanding of the intertwined role of the immune response in atherosclerosis and AD and the implications of these findings for human health.
Collapse
|
29
|
Keeter WC, Ma S, Stahr N, Moriarty AK, Galkina EV. Atherosclerosis and multi-organ-associated pathologies. Semin Immunopathol 2022; 44:363-374. [PMID: 35238952 PMCID: PMC9069968 DOI: 10.1007/s00281-022-00914-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the vascular system that is characterized by the deposition of modified lipoproteins, accumulation of immune cells, and formation of fibrous tissue within the vessel wall. The disease occurs in vessels throughout the body and affects the functions of almost all organs including the lymphoid system, bone marrow, heart, brain, pancreas, adipose tissue, liver, kidneys, and gastrointestinal tract. Atherosclerosis and associated factors influence these tissues via the modulation of local vascular functions, induction of cholesterol-associated pathologies, and regulation of local immune responses. In this review, we discuss how atherosclerosis interferers with functions of different organs via several common pathways and how the disturbance of immunity in atherosclerosis can result in disease-provoking dysfunctions in multiple tissues. Our growing appreciation of the implication of atherosclerosis and associated microenvironmental conditions in the multi-organ pathology promises to influence our understanding of CVD-associated disease pathologies and to provide new therapeutic opportunities.
Collapse
Affiliation(s)
- W Coles Keeter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Shelby Ma
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Natalie Stahr
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Alina K Moriarty
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA
| | - Elena V Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 West Olney Rd, Norfolk, VA, 23507, USA.
| |
Collapse
|
30
|
Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis 2022; 37:67-104. [PMID: 34719771 DOI: 10.1007/s11011-021-00800-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative diseases (NDs) are characterised by progressive dysfunction of synapses, neurons, glial cells and their networks. Neurodegenerative diseases can be classified according to primary clinical features (e.g., dementia, parkinsonism, or motor neuron disease), anatomic distribution of neurodegeneration (e.g., frontotemporal degenerations, extrapyramidal disorders, or spinocerebellar degenerations), or principal molecular abnormalities. The most common neurodegenerative disorders are amyloidosis, tauopathies, a-synucleinopathy, and TAR DNA-binding protein 43 (TDP-43) proteopathy. The protein abnormalities in these disorders have abnormal conformational properties along with altered cellular mechanisms, and they exhibit motor deficit, mitochondrial malfunction, dysfunctions in autophagic-lysosomal pathways, synaptic toxicity, and more emerging mechanisms such as the roles of stress granule pathways and liquid-phase transitions. Finally, for each ND, microglial cells have been reported to be implicated in neurodegeneration, in particular, because the microglial responses can shift from neuroprotective to a deleterious role. Growing experimental evidence suggests that abnormal protein conformers act as seed material for oligomerization, spreading from cell to cell through anatomically connected neuronal pathways, which may in part explain the specific anatomical patterns observed in brain autopsy sample. In this review, we mention the human pathology of select neurodegenerative disorders, focusing on how neurodegenerative disorders (i.e., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis) represent a great healthcare problem worldwide and are becoming prevalent because of the increasing aged population. Despite many studies have focused on their etiopathology, the exact cause of these diseases is still largely unknown and until now with the only available option of symptomatic treatments. In this review, we aim to report the systematic and clinically correlated potential biomarker candidates. Although future studies are necessary for their use in early detection and progression in humans affected by NDs, the promising results obtained by several groups leads us to this idea that biomarkers could be used to design a potential therapeutic approach and preclinical clinical trials for the treatments of NDs.
Collapse
Affiliation(s)
- Nishit Pathak
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Ishi Tandon
- Amity University Jaipur, Rajasthan, Jaipur, Rajasthan, India
| | - Lokesh Agrawal
- Graduate School of Comprehensive Human Sciences, Kansei Behavioural and Brain Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Cao Hongyi
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China
| | - Sanjib Bhattacharyya
- Department of Pharmaceutical Sciences and Chinese Traditional Medicine, Southwest University, Beibei, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
31
|
Gutierrez J, Porras TN, Yoo-Jeong M, Khasiyev F, Igwe KC, Laing KK, Brickman AM, Pavol M, Schnall R. Cerebrovascular Contributions to Neurocognitive Disorders in People Living With HIV. J Acquir Immune Defic Syndr 2021; 88:79-85. [PMID: 34397745 PMCID: PMC8371714 DOI: 10.1097/qai.0000000000002729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/07/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND To investigate a comprehensive array of magnetic resonance imaging (MRI)-based biomarkers of cerebrovascular disease (CVD) in a cohort of people living with HIV (PLWH) and relate these imaging biomarkers to cognition. SETTINGS Cross-sectional, community-based study. METHODS Participants were PLWH in New York City, aged 50 years or older. They underwent a brain magnetic resonance angiography or MRI to ascertain 7 MRI markers of CVD: silent brain infarcts, dilated perivascular spaces, microhemorrhages, white matter hyperintensity volume, white matter fractional anisotropy and mean diffusivity (measures of white matter integrity), and intracranial large artery stenosis. Participants underwent a battery of neurocognitive tests to obtain individual and global cognitive scores representative of various aspects of cognition. RESULTS We included 85 participants (mean age 60 ± 6 years, 48% men, 78% non-Hispanic Black), most of them with well-controlled HIV (75% with CD4 cell count > 200 cells/mm3 and viral load < 400 copies/mL at or near the time of the MRI scan). Silent brain infarcts, intracranial large artery stenosis, and poor white matter integrity were associated with poorer performance in at least one cognitive domain, but the sum of these 3 MRI markers of CVD was associated with lower working memory (B = -0.213, P = 0.028), list learning (B = -0.275, P = 0.019), and global cognition (B = -0.129, P = 0.007). CONCLUSIONS We identified silent brain infarcts, intracranial large artery stenosis, and poor white matter integrity as exposures that may be modifiable and may, therefore, influence cognitive decline. In addition, these MRI markers of CVD may help in identifying PLWH at higher risk of cognitive decline, which may be more amenable to targeted therapies.
Collapse
Affiliation(s)
- Jose Gutierrez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
| | - Tiffany N Porras
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | - Moka Yoo-Jeong
- School of Nursing, Bouvé College of Health Sciences, Northeastern University, Boston, MA
| | - Farid Khasiyev
- Department of Neurology, Saint Louis University, Saint Louis, MI
| | - Kay C Igwe
- Department of Neurology, Columbia University Irving Medical Center, New York, NY
| | - Krystal K Laing
- Department of Neurology, Columbia University Irving Medical Center, New York, NY
| | - Adam M Brickman
- Department of Neurology, Columbia University Irving Medical Center, New York, NY
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY
| | - Marykay Pavol
- Department of Neurology, Columbia University Irving Medical Center, New York, NY
| | - Rebecca Schnall
- School of Nursing, Columbia University Irving Medical Center, New York, NY; and
- Department of Population and Family Health, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
32
|
Maciejewska K, Czarnecka K, Szymański P. A review of the mechanisms underlying selected comorbidities in Alzheimer's disease. Pharmacol Rep 2021; 73:1565-1581. [PMID: 34121170 PMCID: PMC8599320 DOI: 10.1007/s43440-021-00293-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the central nervous system (CNS) leading to mental deterioration and devastation, and eventually a fatal outcome. AD affects mostly the elderly. AD is frequently accompanied by hypercholesterolemia, hypertension, atherosclerosis, and diabetes mellitus, and these are significant risk factors of AD. Other conditions triggered by the progression of AD include psychosis, sleep disorders, epilepsy, and depression. One important comorbidity is Down’s syndrome, which directly contributes to the severity and rapid progression of AD. The development of new therapeutic strategies for AD includes the repurposing of drugs currently used for the treatment of comorbidities. A better understanding of the influence of comorbidities on the pathogenesis of AD, and the medications used in its treatment, might allow better control of disease progression, and more effective pharmacotherapy.
Collapse
Affiliation(s)
- Karolina Maciejewska
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Kamila Czarnecka
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland
| | - Paweł Szymański
- Department of Pharmaceutical Chemistry, Drug Analyses and Radiopharmacy, Faculty of Pharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland.
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St, 01-163, Warsaw, Poland.
| |
Collapse
|
33
|
Duong MT, Nasrallah IM, Wolk DA, Chang CCY, Chang TY. Cholesterol, Atherosclerosis, and APOE in Vascular Contributions to Cognitive Impairment and Dementia (VCID): Potential Mechanisms and Therapy. Front Aging Neurosci 2021; 13:647990. [PMID: 33841127 PMCID: PMC8026881 DOI: 10.3389/fnagi.2021.647990] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are a common cause of cognitive decline, yet limited therapies exist. This cerebrovascular disease results in neurodegeneration via acute, chronic, local, and systemic mechanisms. The etiology of VCID is complex, with a significant impact from atherosclerosis. Risk factors including hypercholesterolemia and hypertension promote intracranial atherosclerotic disease and carotid artery stenosis (CAS), which disrupt cerebral blood flow and trigger ischemic strokes and VCID. Apolipoprotein E (APOE) is a cholesterol and phospholipid carrier present in plasma and various tissues. APOE is implicated in dyslipidemia and Alzheimer disease (AD); however, its connection with VCID is less understood. Few experimental models for VCID exist, so much of the present information has been drawn from clinical studies. Here, we review the literature with a focus on the clinical aspects of atherosclerotic cerebrovascular disease and build a working model for the pathogenesis of VCID. We describe potential intermediate steps in this model, linking cholesterol, atherosclerosis, and APOE with VCID. APOE4 is a minor isoform of APOE that promotes lipid dyshomeostasis in astrocytes and microglia, leading to chronic neuroinflammation. APOE4 disturbs lipid homeostasis in macrophages and smooth muscle cells, thus exacerbating systemic inflammation and promoting atherosclerotic plaque formation. Additionally, APOE4 may contribute to stromal activation of endothelial cells and pericytes that disturb the blood-brain barrier (BBB). These and other risk factors together lead to chronic inflammation, atherosclerosis, VCID, and neurodegeneration. Finally, we discuss potential cholesterol metabolism based approaches for future VCID treatment.
Collapse
Affiliation(s)
- Michael Tran Duong
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ilya M Nasrallah
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David A Wolk
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Ta-Yuan Chang
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
34
|
Alzheimer's Disease and Vascular Aging: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:942-951. [PMID: 32130930 PMCID: PMC8046164 DOI: 10.1016/j.jacc.2019.10.062] [Citation(s) in RCA: 247] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/27/2019] [Indexed: 01/23/2023]
Abstract
Alzheimer’s disease, the leading cause of dementia in the elderly, is a neurodegenerative condition characterized by accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, age-related vascular changes accompany or even precede the development of Alzheimer’s pathology, raising the possibility that they may have a pathogenic role. This review provides an appraisal of the alterations in cerebral and systemic vasculature, the heart, and hemostasis that occur in Alzheimer’s disease and their relationships to cognitive impairment. Although the molecular pathogenesis of these alterations remains to be defined, amyloid-β is a likely contributor in the brain as in the heart. Collectively, the evidence suggests that vascular pathology is a likely pathogenic contributor to age-related dementia, including Alzheimer’s disease, inextricably linked to disease onset and progression. Consequently, the contribution of vascular factors should be considered in preventive, diagnostic, and therapeutic approaches to address one of the major health challenges of our time.
Collapse
|
35
|
Fluid Candidate Biomarkers for Alzheimer's Disease: A Precision Medicine Approach. J Pers Med 2020; 10:jpm10040221. [PMID: 33187336 PMCID: PMC7712586 DOI: 10.3390/jpm10040221] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
A plethora of dynamic pathophysiological mechanisms underpins highly heterogeneous phenotypes in the field of dementia, particularly in Alzheimer's disease (AD). In such a faceted scenario, a biomarker-guided approach, through the implementation of specific fluid biomarkers individually reflecting distinct molecular pathways in the brain, may help establish a proper clinical diagnosis, even in its preclinical stages. Recently, ultrasensitive assays may detect different neurodegenerative mechanisms in blood earlier. ß-amyloid (Aß) peptides, phosphorylated-tau (p-tau), and neurofilament light chain (NFL) measured in blood are gaining momentum as candidate biomarkers for AD. P-tau is currently the more convincing plasma biomarker for the diagnostic workup of AD. The clinical role of plasma Aβ peptides should be better elucidated with further studies that also compare the accuracy of the different ultrasensitive techniques. Blood NFL is promising as a proxy of neurodegeneration process tout court. Protein misfolding amplification assays can accurately detect α-synuclein in cerebrospinal fluid (CSF), thus representing advancement in the pathologic stratification of AD. In CSF, neurogranin and YKL-40 are further candidate biomarkers tracking synaptic disruption and neuroinflammation, which are additional key pathophysiological pathways related to AD genesis. Advanced statistical analysis using clinical scores and biomarker data to bring together individuals with AD from large heterogeneous cohorts into consistent clusters may promote the discovery of pathophysiological causes and detection of tailored treatments.
Collapse
|
36
|
Shafi O. Switching of vascular cells towards atherogenesis, and other factors contributing to atherosclerosis: a systematic review. Thromb J 2020; 18:28. [PMID: 33132762 PMCID: PMC7592591 DOI: 10.1186/s12959-020-00240-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Onset, development and progression of atherosclerosis are complex multistep processes. Many aspects of atherogenesis are not yet properly known. This study investigates the changes in vasculature that contribute to switching of vascular cells towards atherogenesis, focusing mainly on ageing. Methods Databases including PubMed, MEDLINE and Google Scholar were searched for published articles without any date restrictions, involving atherogenesis, vascular homeostasis, aging, gene expression, signaling pathways, angiogenesis, vascular development, vascular cell differentiation and maintenance, vascular stem cells, endothelial and vascular smooth muscle cells. Results Atherogenesis is a complex multistep process that unfolds in a sequence. It is caused by alterations in: epigenetics and genetics, signaling pathways, cell circuitry, genome stability, heterotypic interactions between multiple cell types and pathologic alterations in vascular microenvironment. Such alterations involve pathological changes in: Shh, Wnt, NOTCH signaling pathways, TGF beta, VEGF, FGF, IGF 1, HGF, AKT/PI3K/ mTOR pathways, EGF, FOXO, CREB, PTEN, several apoptotic pathways, ET - 1, NF-κB, TNF alpha, angiopoietin, EGFR, Bcl - 2, NGF, BDNF, neurotrophins, growth factors, several signaling proteins, MAPK, IFN, TFs, NOs, serum cholesterol, LDL, ephrin, its receptor pathway, HoxA5, Klf3, Klf4, BMPs, TGFs and others.This disruption in vascular homeostasis at cellular, genetic and epigenetic level is involved in switching of the vascular cells towards atherogenesis. All these factors working in pathologic manner, contribute to the development and progression of atherosclerosis. Conclusion The development of atherosclerosis involves the switching of gene expression towards pro-atherogenic genes. This happens because of pathologic alterations in vascular homeostasis. When pathologic alterations in epigenetics, genetics, regulatory genes, microenvironment and vascular cell biology accumulate beyond a specific threshold, then the disease begins to express itself phenotypically. The process of biological ageing is one of the most significant factors in this aspect as it is also involved in the decline in homeostasis, maintenance and integrity.The process of atherogenesis unfolds sequentially (step by step) in an interconnected loop of pathologic changes in vascular biology. Such changes are involved in 'switching' of vascular cells towards atherosclerosis.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
37
|
Kang KM, Byun MS, Lee JH, Yi D, Choi HJ, Lee E, Lee Y, Lee JY, Kim YK, Sohn BK, Sohn CH, Lee DY. Association of carotid and intracranial stenosis with Alzheimer's disease biomarkers. ALZHEIMERS RESEARCH & THERAPY 2020; 12:106. [PMID: 32912336 PMCID: PMC7488394 DOI: 10.1186/s13195-020-00675-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022]
Abstract
Background To clarify whether atherosclerosis of the carotid and intracranial arteries is related to Alzheimer’s disease (AD) pathology in vivo, we investigated the associations of carotid and intracranial artery stenosis with cerebral beta-amyloid (Aβ) deposition and neurodegeneration in middle- and old-aged individuals. Given different variations of the pathologies between cognitive groups, we focused separately on cognitively normal (CN) and cognitively impaired (CI) groups. Methods A total of 281 CN and 199 CI (mild cognitive impairment and AD dementia) subjects underwent comprehensive clinical assessment, [11C] Pittsburgh compound B-positron emission tomography, and magnetic resonance (MR) imaging including MR angiography. We evaluated extracranial carotid and intracranial arteries for the overall presence, severity (i.e., number and degree of narrowing), and location of stenosis. Results We found no associations between carotid and intracranial artery stenosis and cerebral Aβ burden in either the CN or the CI group. In terms of neurodegeneration, exploratory univariable analyses showed associations between the presence and severity of stenosis and regional neurodegeneration biomarkers (i.e., reduced hippocampal volume [HV] and cortical thickness in the AD-signature regions) in both the CN and CI groups. In confirmatory multivariable analyses controlling for demographic covariates and diagnosis, the association between number of stenotic intracranial arteries ≥ 2 and reduced HV in the CI group remained significant. Conclusions Neither carotid nor intracranial artery stenosis appears to be associated with brain Aβ burden, while intracranial artery stenosis is related to amyloid-independent neurodegeneration, particularly hippocampal atrophy.
Collapse
Affiliation(s)
- Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, National Center for Mental Health, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Hye Jeong Choi
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Eunjung Lee
- Department of Radiology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Younghwa Lee
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Bo Kyung Sohn
- Department of Psychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea. .,Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea. .,Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | | |
Collapse
|
38
|
Fujiyoshi A, Suri MFK, Alonso A, Selvin E, Chu H, Guallar E, Qiao Y, Zhang Y, Wasserman BA, Folsom AR. Hyperglycemia, duration of diabetes, and intracranial atherosclerotic stenosis by magnetic resonance angiography: The ARIC-NCS study. J Diabetes Complications 2020; 34:107605. [PMID: 32600893 PMCID: PMC8285273 DOI: 10.1016/j.jdiacomp.2020.107605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 01/31/2023]
Abstract
AIMS The association of hyperglycemia and duration of diabetes with intracranial atherosclerotic stenosis (ICAS) in the general population is not well documented. We examined whether elevated glucose and longer diabetes duration is independently associated with ICAS in a community-based sample. METHODS We cross-sectionally analyzed 1644 participants (age 67-90 years) of the Atherosclerosis Risk in Communities Study who underwent cerebrovascular magnetic resonance angiography in 2011-13. We applied multivariable ordinal logistic regression to evaluate the association of ICAS category ("no stenosis", "stenosis <50%", or "stenosis ≥50%") with glucose or diabetes duration (<10, 10 to 20, and ≥20 years). We also obtained the corresponding odds ratios applying inverse-probability weighting to account for potential selection bias due to attrition. RESULTS Compared to non-diabetic participants in the lowest glucose quartile, the weighted odds ratios (95% confidence interval) of higher ICAS category were 1.88 (1.18, 3.00) and 2.01 (1.08, 3.72) for non-diabetic and diabetic participants in the corresponding highest glucose quartile, respectively. We observed significant positive trends of ICAS across diabetes duration categories in unweighted, but not in weighted, analyses. CONCLUSIONS Hyperglycemia and longer duration of diabetes were independently associated with ICAS, suggesting the importance of maintaining glycemic control to prevent stroke.
Collapse
Affiliation(s)
- Akira Fujiyoshi
- Department of Hygiene, Wakayama Medical University, Wakayama, Japan.
| | - M Fareed K Suri
- Department of Neurology, University of Minnesota, MN, United States of America
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Elizabeth Selvin
- Departments of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Haitao Chu
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| | - Eliseo Guallar
- Departments of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Ye Qiao
- The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Yiyi Zhang
- Departments of Epidemiology and the Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Bruce A Wasserman
- The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Aaron R Folsom
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
39
|
Hocker JR, Lerner M, Lightfoot SA, Peyton MD, Thompson JL, Deb S, Reinersman M, Hanas RJ, Postier RG, Edil BH, Burkhart HM, Hanas JS. Serum discrimination and phenotype assessment of coronary artery disease patents with and without type 2 diabetes prior to coronary artery bypass graft surgery. PLoS One 2020; 15:e0234539. [PMID: 32756554 PMCID: PMC7527241 DOI: 10.1371/journal.pone.0234539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 05/12/2020] [Indexed: 11/18/2022] Open
Abstract
Diabetes Mellitus (DM) accelerates coronary artery disease (CAD) and atherosclerosis, the causes of most heart attacks. The biomolecules involved in these inter-related disease processes are not well understood. This study analyzes biomolecules in the sera of patients with CAD, with and without type (T) 2DM, who are about to undergo coronary artery bypass graft (CABG) surgery. The goal is to develop methodology to help identify and monitor CAD patients with and without T2DM, in order to better understand these phenotypes and to glean relationships through analysis of serum biomolecules. Aorta, fat, muscle, and vein tissues from CAD T2DM patients display diabetic-related histologic changes (e.g., lipid accumulation, fibrosis, loss of cellularity) when compared to non-diabetic CAD patients. The patient discriminatory methodology utilized is serum biomolecule mass profiling. This mass spectrometry (MS) approach is able to distinguish the sera of a group of CAD patients from controls (p value 10−15), with the CAD group containing both T2DM and non-diabetic patients. This result indicates the T2DM phenotype does not interfere appreciably with the CAD determination versus control individuals. Sera from a group of T2DM CAD patients however are distinguishable from non-T2DM CAD patients (p value 10−8), indicating it may be possible to examine the T2DM phenotype within the CAD disease state with this MS methodology. The same serum samples used in the CAD T2DM versus non-T2DM binary group comparison were subjected to MS/MS peptide structure analysis to help identify potential biochemical and phenotypic changes associated with CAD and T2DM. Such peptide/protein identifications could lead to improved understanding of underlying mechanisms, additional biomarkers for discriminating and monitoring these disease conditions, and potential therapeutic targets. Bioinformatics/systems biology analysis of the peptide/protein changes associated with CAD and T2DM suggested cell pathways/systems affected include atherosclerosis, DM, fibrosis, lipogenesis, loss of cellularity (apoptosis), and inflammation.
Collapse
Affiliation(s)
- James R. Hocker
- Department of Biochemistry and Molecular Biology The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Megan Lerner
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Stan A. Lightfoot
- Department of Medicine The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Marvin D. Peyton
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jess L. Thompson
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Subrato Deb
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Mathew Reinersman
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - R. Jane Hanas
- Department of Biochemistry and Molecular Biology The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Russel G. Postier
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Barish H. Edil
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Harold M. Burkhart
- Department of Surgery The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jay S. Hanas
- Department of Biochemistry and Molecular Biology The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
40
|
Eglit GML, Weigand AJ, Nation DA, Bondi MW, Bangen KJ. Hypertension and Alzheimer's disease: indirect effects through circle of Willis atherosclerosis. Brain Commun 2020; 2:fcaa114. [PMID: 33543127 PMCID: PMC7846096 DOI: 10.1093/braincomms/fcaa114] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Hypertension is common among older adults and is believed to increase susceptibility to Alzheimer's disease, but mechanisms underlying this relationship are unclear. Hypertension also promotes circle of Willis atherosclerosis, which contributes to cerebral hypoperfusion and arterial wall stiffening, two potential mechanisms linking hypertension to Alzheimer's disease. To examine the role of circle of Willis atherosclerosis in the association between hypertension and Alzheimer's disease neuropathology, we analysed post-mortem neuropathological data on 2198 decedents from the National Alzheimer's Coordinating Center database [mean (standard deviation) age at last visit 80.51 (1.95) and 47.1% female] using joint simultaneous (i.e. mediation) modelling. Within the overall sample and among Alzheimer's dementia decedents, hypertension was indirectly associated with increased neuritic plaques and neurofibrillary tangles through its association with circle of Willis atherosclerosis. Similar indirect effects were observed for continuous measures of systolic and diastolic blood pressure. These results suggest that hypertension may promote Alzheimer's disease pathology indirectly through intracranial atherosclerosis by limiting cerebral blood flow and/or dampening perivascular clearance. Circle of Willis atherosclerosis may be an important point of convergence between vascular risk factors, cerebrovascular changes and Alzheimer's disease neuropathology.
Collapse
Affiliation(s)
- Graham M L Eglit
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Alexandra J Weigand
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA
| | - Daniel A Nation
- Department of Psychological Science, University of California, Irvine, Irvine, CA 92697, USA
- Institute for Memory Disorders and Neurological Impairments, University of California, Irvine, Irvine, CA 92697, USA
| | - Mark W Bondi
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| | - Katherine J Bangen
- Veteran Affairs San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
41
|
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: 108] [Impact Index Per Article: 21.6] [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
|
42
|
Gamma glutamyltransferase and risk of dementia in prediabetes and diabetes. Sci Rep 2020; 10:6800. [PMID: 32322005 PMCID: PMC7176727 DOI: 10.1038/s41598-020-63803-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Diabetes is associated with cognitive impairment and greater risk for dementia, but the role of gamma-glutamyltransferase (γ-GT) in dementia has not been elucidated. We determined incident dementia including Alzheimer’s disease and vascular dementia, analyzing data from participants aged 40 years or older in the National Health Insurance Database, collected by the National Health Insurance Service in Korea, from January 2009 to December 2015. During a median follow-up of 7.6 years, 272,657 participants were diagnosed as having dementia. Higher serum γ-GT was associated with increased risk of dementia (HR = 1.22, 95% CI = 1.20–1.24), and had a strong positive association with early onset dementia (HR = 1.32, 95% CI = 1.24–1.40). An additive impact of higher γ-GT on dementia was observed regardless of glycemic status, and prevalent diabetes with the highest γ-GT quartile had a 1.8-fold increased dementia risk (HR = 1.82, 95% CI = 1.78–1.85). This effect of γ-GT concentration in diabetes was more prominent in individuals with vascular dementia (HR = 1.94, 95% CI = 1.84–2.04). In subgroup analysis, young age, male sex, and relatively healthy subjects with a higher γ-GT quartile had more increased dementia risk. In conclusion, γ-GT concentration as well as glycemic status could be a future risk factor for dementia in the general population.
Collapse
|
43
|
Gutierrez J, Byrd D, Yin MT, Morgello S. Relationship Between Brain Arterial Pathology and Neurocognitive Performance Among Individuals With Human Immunodeficiency Virus. Clin Infect Dis 2020; 68:490-497. [PMID: 30107467 DOI: 10.1093/cid/ciy501] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/11/2018] [Indexed: 12/24/2022] Open
Abstract
Background Human immunodeficiency virus-positive (HIV+) individuals have higher rates of cognitive impairment and cerebrovascular disease compared with uninfected populations. We hypothesize that cerebrovascular disease, specifically brain large artery disease, may play a role in HIV-associated neurocognitive disorders (HAND). Methods Participants (N = 94) in the Manhattan HIV Brain Bank study were followed on average 32 ± 33 months with repeated neuropsychological examinations until death. We used five cognitive domains (motor, processing speed, working memory, verbal fluency, and executive functioning) to assess ante mortem performance. We quantified the diameter of the lumen and arterial wall thickness obtained during autopsy. The diagnoses of HAND were attributed using the American Academy of Neurology nosology. We used generalized linear mixed model to account for repeated measures, follow-up time, and codependence between arteries. Models were adjusted for demographics, viral loads, CD4 counts, history of opportunistic infections, and vascular risks. Results We included 94 HIV+ individuals (mean age 56 ± 8.3, 68% men, 54% African American). In adjusted models, there was an association between arterial wall thickness and global cognitive score (B = -0.176, P value = .03), processing speed (B = -0.175, P = .05), and verbal fluency (B = -0.253, P = .02). Participants with incident or worsening HAND had thicker brain arterial walls (B = 0.523 ± 0.234, P = .03) and smaller arterial lumen (B = -0.633 ± 0.252, P = .01). Conclusions We report here a novel association between brain arterial wall thickening and poorer ante mortem cognitive performance and diagnosis of incident or worsening HAND at death. Strategies to preserve the arterial lumen or to prevent wall thickening may impact HAND.
Collapse
Affiliation(s)
- Jose Gutierrez
- Department of Neurology, Stroke Division, New York Presbyterian/Columbia University Medical Center, New York
| | - Desiree Byrd
- Departments of Neurology and Pathology, Mount Sinai School of Medicine, New York Presbyterian/Columbia University Medical Center, New York
| | - Michael T Yin
- Department of Medicine, Infectious Disease Division, New York Presbyterian/Columbia University Medical Center, New York
| | - Susan Morgello
- Departments of Neurology and Pathology, Mount Sinai School of Medicine, New York Presbyterian/Columbia University Medical Center, New York
| |
Collapse
|
44
|
Xia C, Vonder M, Sidorenkov G, Oudkerk M, de Groot JC, van der Harst P, de Bock GH, De Deyn PP, Vliegenthart R. The Relationship of Coronary Artery Calcium and Clinical Coronary Artery Disease with Cognitive Function: A Systematic Review and Meta-Analysis. J Atheroscler Thromb 2020; 27:934-958. [PMID: 32062643 PMCID: PMC7508729 DOI: 10.5551/jat.52928] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIM Coronary artery disease (CAD) and cognitive impairment are common in the elderly, with evidence for shared risk factors and pathophysiological processes. The coronary artery calcium (CAC) score is a marker of subclinical CAD, which may allow early detection of individuals prone to cognitive decline. Prior studies on associations of CAC and clinical CAD with cognitive impairment had discrepant results. This systematic review aims to evaluate the association of (sub)clinical CAD with cognitive function, cognitive decline, and diagnosis of mild cognitive impairment (MCI) or dementia. METHODS A systematic search was conducted in MEDLINE, Embase, and Web of Science until February 2019, supplemented with citations tracking. Two reviewers independently screened studies and extracted information including odds ratios (ORs) and hazard ratios (HRs). RESULTS Forty-six studies, 10 on CAC and 36 on clinical CAD, comprising 1,248,908 participants were included in the systematic review. Studies about associations of (sub)clinical CAD with cognitive function and cognitive decline had heterogeneous methodology and inconsistent findings. Two population-based studies investigated the association between CAC and risk of dementia over 6-12.2 years using different CAC scoring methods. Both found a tendency toward higher risk of dementia as CAC severity increased. Meta-analysis in 15 studies (663,250 individuals) showed an association between CAD and MCI/dementia (pooled OR 1.32, 95%CI 1.17-1.48) with substantial heterogeneity (I2=87.0%, p<0.001). Pooled HR of CAD for incident MCI/dementia over 3.2-25.5 years in six longitudinal studies (70,060 individuals) was 1.51 (95%CI 1.24-1.85), with low heterogeneity (I2=14.1%, p=0.32). Sensitivity analysis did not detect any study that was of particular influence on the pooled OR or HR. CONCLUSIONS Limited evidence suggests the CAC score is associated with risk of dementia. In clinical CAD, risk of MCI and dementia is increased by 50%, as supported by stronger evidence.
Collapse
Affiliation(s)
- Congying Xia
- University of Groningen, University Medical Center Groningen, Department of Radiology
| | - Marleen Vonder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology
| | - Grigory Sidorenkov
- University of Groningen, University Medical Center Groningen, Department of Epidemiology
| | | | - Jan Cees de Groot
- University of Groningen, University Medical Center Groningen, Department of Radiology
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology
| | - Geertruida H de Bock
- University of Groningen, University Medical Center Groningen, Department of Epidemiology
| | - Peter Paul De Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Alzheimer Center Groningen
| | | |
Collapse
|
45
|
Fungwe TV, Ngwa JS, Ntekim OE, Allard JS, Nadarajah S, Wolday S, Ogunlana OO, Johnson SP, Hughes K, Larbi D, Gillum RF, Obisesan TO. Exercise Training Induced Changes In Nuclear Magnetic Resonance-Measured Lipid Particles In Mild Cognitively Impaired Elderly African American Volunteers: A Pilot Study. Clin Interv Aging 2019; 14:2115-2123. [PMID: 31824142 PMCID: PMC6900999 DOI: 10.2147/cia.s195878] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/02/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose Poor cardiorespiratory fitness (CRF) is linked to cognitive deterioration, but its effects on lipid heterogeneity and functional properties in older African American (AA) subjects with mild cognitive impairment (MCI) need elucidation. This study determined whether exercise training-induced changes in blood lipid particle sizes (LPS) were associated with CRF determined by VO2Max in elderly AAs with MCI. Given the pivotal role of brain-derived neurotrophic factor (BDNF) on glucose metabolism, and therefore, “diabetic dyslipidemia”, we also determined whether changes in LPS were associated with the levels of serum BDNF. Methods This analysis included 17 of the 29 randomized elderly AAs with MCI who had NMR data at baseline and after a 6-month training. We used Generalized Linear Regression (GLM) models to examine cardiorespiratory fitness (VO2Max) effects on training-induced change in LPS in the stretch and aerobic groups. Additionally, we determined whether the level of BDNF influenced change in LPS. Results Collectively, mean VO2Max (23.81±6.17) did not differ significantly between aerobic and stretch groups (difference=3.17±3.56, P=0.495). Training-related changes in very low-density lipoprotein, chylomicrons, and total low-density lipoprotein (LDL) particle sizes correlated significantly with VO2Max, but not after adjustment for age and gender. However, increased VO2Max significantly associated with reduced total LDL particle size after similar adjustments (P = 0.046). While stretch exercise associated with increased protective large high-density lipoprotein particle size, the overall effect was not sustained following adjustments for gender and age. However, changes in serum BDNF were associated with changes in triglyceride and cholesterol transport particle sizes (P < 0.051). Conclusion Promotion of stretch and aerobic exercise to increase CRF in elderly AA volunteers with MCI may also promote beneficial changes in lipoprotein particle profile. Because high BDNF concentration may reduce CVD risk, training-related improvements in BDNF levels are likely advantageous. Large randomized studies are needed to confirm our observations and to further elucidate the role for exercise therapy in reducing CVD risk in elderly AAs with MCI.
Collapse
Affiliation(s)
- Thomas V Fungwe
- Department of Nutritional Sciences, School of Nursing and Allied Health Sciences, Howard University, Washington, DC, USA
| | - Julius S Ngwa
- Division of Cardiology, Department of Medicine, Howard University, Washington, DC, USA
| | - Oyonumo E Ntekim
- Department of Nutritional Sciences, School of Nursing and Allied Health Sciences, Howard University, Washington, DC, USA
| | - Joanne S Allard
- Division of Cardiology, Department of Physiology and Biophysics, Howard University, Washington, DC, USA
| | - Sheeba Nadarajah
- Division of Nursing, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, USA
| | - Saba Wolday
- Division of Geriatrics, Department of Medicine and Clinical, Howard University Hospital, Washington, DC, USA
| | - Oludolapo O Ogunlana
- Division of Geriatrics, Department of Medicine and Clinical, Howard University Hospital, Washington, DC, USA
| | - Steven P Johnson
- Division of Geriatrics, Department of Medicine and Clinical, Howard University Hospital, Washington, DC, USA
| | - Kakra Hughes
- Department of Surgery, Howard University College of Medicine, Washington, DC, USA
| | - Daniel Larbi
- Division of Geriatrics, Department of Medicine and Clinical, Howard University Hospital, Washington, DC, USA
| | - Richard F Gillum
- Department of Internal Medicine, Howard University Hospital, Washington, DC, USA
| | - Thomas O Obisesan
- Division of Geriatrics, Department of Medicine and Clinical, Howard University Hospital, Washington, DC, USA
| |
Collapse
|
46
|
Declerck K, Vanden Berghe W. Characterization of Blood Surrogate Immune-Methylation Biomarkers for Immune Cell Infiltration in Chronic Inflammaging Disorders. Front Genet 2019; 10:1229. [PMID: 31827492 PMCID: PMC6890858 DOI: 10.3389/fgene.2019.01229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/06/2019] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) and atherosclerosis are both chronic age- and inflammation-dependent diseases. In addition, atherosclerosis is frequently observed in AD patients indicating common involvement of vascular components in both disease etiologies. Recently, epigenome-wide association studies have identified epigenetic alterations, and in particularly DNA methylation changes for both disorders. We hypothesized the existence of a common DNA methylation profile in atherosclerosis and AD which may be valuable as a blood-based DNA methylation inflammaging biomarker. Using publicly available 450k Illumina methylation datasets, we identified a co-methylation network associated with both atherosclerosis and AD in whole blood samples. This methylation profile appeared to indicate shifts in blood immune cell type distribution. Remarkably, similar methylation changes were also detected in disease tissues, including AD brain tissues, atherosclerotic plaques, and tumors and were found to correlate with immune cell infiltration. In addition, this immune-related methylation profile could also be detected in other inflammaging diseases, including Parkinson’s disease and obesity, but not in multiple sclerosis, schizophrenia, and osteoporosis. In conclusion, we identified a blood-based immune-related DNA methylation signature in multiple inflammaging diseases associated with changes in blood immune cell counts and predictive for immune cell infiltration in diseased tissues. In addition to epigenetic clock measurements, this immune-methylation signature may become a valuable blood-based biomarker to prevent chronic inflammatory disease development or monitor lifestyle intervention strategies which promote healthy aging.
Collapse
Affiliation(s)
- Ken Declerck
- Laboratory of Protein Chemistry, Proteomics, and Epigenetic Signaling (PPES), Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, Antwerp University, Antwerp, Belgium
| | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics, and Epigenetic Signaling (PPES), Department of Biomedical Sciences, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, Antwerp University, Antwerp, Belgium
| |
Collapse
|
47
|
Gustavsson AM, van Westen D, Stomrud E, Engström G, Nägga K, Hansson O. Midlife Atherosclerosis and Development of Alzheimer or Vascular Dementia. Ann Neurol 2019; 87:52-62. [PMID: 31721283 PMCID: PMC6973178 DOI: 10.1002/ana.25645] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 10/03/2019] [Accepted: 11/10/2019] [Indexed: 12/28/2022]
Abstract
Objective To investigate whether midlife atherosclerosis is associated with different dementia subtypes and related underlying pathologies. Methods Participants comprised the cardiovascular cohort of the Swedish prospective population‐based Malmö Diet and Cancer Study (N = 6,103). Carotid plaques and intima media thickness (IMT) were measured at baseline (1991–1994). Dementia incidence until 2014 was obtained from national registers. Diagnoses were reviewed and validated in medical records. In a cognitively unimpaired subcohort (n = 330), β‐amyloid42 and tau were quantified in cerebrospinal fluid (CSF), and white matter hyperintensity volume, lacunar infarcts, and cerebral microbleeds were estimated on magnetic resonance imaging (2009–2015). Results During 20 years of follow‐up, 462 individuals developed dementia (mean age at baseline = 57.5 ± 5.9 years, 58% women). Higher IMT in midlife was associated with an increased hazard ratio (HR) of all‐cause dementia (adjusted HR = 1.14 [95% confidence interval (CI) = 1.03–1.26]) and vascular dementia (adjusted HR = 1.32 [95% CI = 1.10–1.57]) but not Alzheimer disease (AD) dementia (adjusted HR = 0.95 [95% CI = 0.77–1.17]). Carotid plaques were associated with vascular dementia when assessed as a 3‐graded score (adjusted HR = 1.90 [95% CI = 1.07–3.38]). In the cognitively unimpaired subcohort (53.8 ± 4.6 years at baseline, 60% women), higher IMT in midlife was associated with development of small vessel disease (adjusted odds ratio [OR] = 1.47 [95% CI = 1.05–2.06]) but not significantly with abnormal CSF AD biomarkers (adjusted OR = 1.28 [95% CI = 0.87–1.90] for Aβ42 and 1.35 [95% CI = 0.86–2.13] for Aβ42/p‐tau). Carotid plaques revealed no significant association with any of the underlying brain pathologies. Interpretation Our findings support an association between midlife atherosclerosis and development of vascular dementia and cerebral small vessel disease but not between atherosclerosis and subsequent AD dementia or AD pathology. ANN NEUROL 2020;87:52–62
Collapse
Affiliation(s)
- Anna-Märta Gustavsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| | - Danielle van Westen
- Diagnostic Radiology, Department of Clinical Sciences Lund, Lund University, Lund
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| | - Gunnar Engström
- Cardiovascular Epidemiology, Department of Clinical Sciences Malmö, Lund University, Malmö
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Department of Acute Internal Medicine and Geriatrics, Linköping University, Linköping, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö.,Memory Clinic, Skåne University Hospital, Malmö
| |
Collapse
|
48
|
Martins AH, Zayas-Santiago A, Ferrer-Acosta Y, Martinez-Jimenez SM, Zueva L, Diaz-Garcia A, Inyushin M. Accumulation of Amyloid Beta (Aβ) Peptide on Blood Vessel Walls in the Damaged Brain after Transient Middle Cerebral Artery Occlusion. Biomolecules 2019; 9:biom9080350. [PMID: 31398804 PMCID: PMC6723874 DOI: 10.3390/biom9080350] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/03/2019] [Accepted: 08/03/2019] [Indexed: 02/06/2023] Open
Abstract
It is well known that amyloid beta (Aβ) peptides are generated in blood vessels, released into the brain during thrombosis, and temporarily accumulate in this organ after injury. Here we demonstrate that 24 h after transient middle cerebral artery occlusion (tMCAO), one of the standard models of focal ischemic stroke, Aβ peptide accumulates in the brain, concentrating on the blood vessel walls. Because Aβ oligomers are known to induce significant damage to brain cells, they act as an additional damaging factor during ischemic stroke. Considering that they have been shown to form ion channels in cells, affecting osmotic balance, we used an Aβ peptide channel blocker, tromethamine (2-amino-2-(hydroxymethyl) propane-1,3-diol), to prevent this additional injury. Tromethamine injected 0.1 g/100 g body weight intraperitoneally at 5 min before tMCAO decreased water content in the damaged hemisphere, as measured by dry brain weight. Congo red staining, which binds only to Aβ oligomer plaques (amyloid), showed that there was no significant presence of plaques. Therefore, we suggest that Aβ peptide oligomers are responsible for some of the brain damage during stroke and that blockage of the ion channels that they form could be beneficial in treating this complex neurological syndrome.
Collapse
Affiliation(s)
- Antonio Henrique Martins
- Pharmacology and Toxicology Department, University of Puerto Rico, Medical Sciences Campus, Guillermo Arbona, Área de Centro Médico Río Piedras, PR 00935, USA
| | - Astrid Zayas-Santiago
- Department of Physiology, Universidad Central del Caribe Ave. Laurel #100, Santa Juanita, Bayamón, PR 00956, USA
| | - Yancy Ferrer-Acosta
- Department of Neuroscience, Universidad Central del Caribe Ave. Laurel #U26, Santa Juanita, Bayamón, PR 00956, USA
| | - Solianne M Martinez-Jimenez
- Department of Neuroscience, Universidad Central del Caribe Ave. Laurel #U26, Santa Juanita, Bayamón, PR 00956, USA
| | - Lidia Zueva
- Department of Physiology, Universidad Central del Caribe Ave. Laurel #100, Santa Juanita, Bayamón, PR 00956, USA
| | - Amanda Diaz-Garcia
- Department of Physiology, Universidad Central del Caribe Ave. Laurel #100, Santa Juanita, Bayamón, PR 00956, USA
| | - Mikhail Inyushin
- Department of Physiology, Universidad Central del Caribe Ave. Laurel #100, Santa Juanita, Bayamón, PR 00956, USA.
| |
Collapse
|
49
|
Inyushin M, Zayas-Santiago A, Rojas L, Kucheryavykh Y, Kucheryavykh L. Platelet-generated amyloid beta peptides in Alzheimer's disease and glaucoma. Histol Histopathol 2019; 34:843-856. [PMID: 30945258 PMCID: PMC6667289 DOI: 10.14670/hh-18-111] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amyloid beta (Aβ) peptides have been implicated in both Alzheimer's disease (AD) and glaucoma and have been shown to be the key etiological factor in these dangerous health complications. On the other hand, it is well known that Aβ peptide can be generated from its precursor protein and massively released from the blood to nearby tissue upon the activation of platelets due to their involvement in innate immunity and inflammation processes. Here we review evidence about the development of AD and glaucoma neuronal damage showing their dependence on platelet count and activation. The correlation between the effect on platelet count and the effectiveness of anti-AD and anti-glaucoma therapies suggest that platelets may be an important player in these diseases.
Collapse
Affiliation(s)
- Mikhail Inyushin
- School of Medicine, Universidad Central del Caribe (UCC), PR, USA.
| | | | - Legier Rojas
- School of Medicine, Universidad Central del Caribe (UCC), PR, USA
| | | | | |
Collapse
|
50
|
Magge SN, Zemel BS, Pipan ME, Gidding SS, Kelly A. Cardiometabolic Risk and Body Composition in Youth With Down Syndrome. Pediatrics 2019; 144:peds.2019-0137. [PMID: 31315916 PMCID: PMC6855833 DOI: 10.1542/peds.2019-0137] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/17/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Whether BMI captures adiposity and cardiometabolic risk in Down syndrome (DS), a condition associated with obesity, short stature, and altered body proportions, is not known. We compared cardiometabolic risk measures in youth with DS and typically developing matched controls. METHODS Youth with (n = 150) and without (n = 103) DS of comparable age (10-20 years), sex, race, ethnicity, and BMI percentile underwent whole-body dual-energy X-ray absorptiometry, fasting glucose, insulin, lipids, lipoprotein particles, inflammatory factors, and when BMI percentile ≥85, an oral glucose tolerance test. RESULTS Sixty-four percent of youth with DS had BMI percentile ≥85. Among these, no difference in glucose, insulin, or insulin resistance was detected, but prediabetes was more prevalent with DS (26.4% vs 10.3%; P = .025) after adjustment for demographics, pubertal status, and BMI z score (odds ratio = 3.2; P = .026). Among all participants, those with DS had higher low-density lipoprotein cholesterol (median 107 [interquartile range 89-128] vs 88.5 [79-103] mg/dL; P < .00005), triglycerides (89.5 [73-133] vs 71.5 [56-104] mg/dL; P < .00005), non-high-density lipoprotein cholesterol (non-HDL-C; 128 [104-153] vs 107 [92-123] mg/dL; P < .00005), and triglycerides/HDL-C (2.2 [1.6-3.4] vs 1.7 [1.1-2.5] mg/dL; P = .0003) and lower levels of HDL-C (41 [36.5-47] vs 45 [37-53] mg/dL; P = .012). DS youth had higher high-sensitivity C-reactive protein, interleukin-6, small low-density lipoprotein particles (LDL-P), and total LDL-P, but similar LDL-P size. Youth with DS had less visceral fat (VFAT), fat mass, and lean mass for BMI z score, but greater VFAT at higher fat mass. However, VFAT did not fully explain the increased prevalence of dyslipidemia or prediabetes in youth with DS. CONCLUSIONS Despite similar insulin resistance, youth with DS had greater prevalence of dyslipidemia and prediabetes than typically developing youth, which was not fully explained by VFAT.
Collapse
Affiliation(s)
- Sheela N. Magge
- Division of Endocrinology and Diabetes, School of Medicine and Health Sciences, The George Washington University and Clinical and Translational Science Institute at Children’s National, Children’s Research Institute, Children’s National Health System, Washington, District of Columbia; Divisions of
| | - Babette S. Zemel
- Gastroenterology, Hepatology, and Nutrition,,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| | - Mary E. Pipan
- Developmental Behavioral Pediatrics, and,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| | | | - Andrea Kelly
- Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania;,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania; and
| |
Collapse
|