1
|
McFadden J, Matthews J, Scott L, Herholz K, Dickie B, Haroon H, Sparasci O, Ahmed S, Kyrtata N, Parker GJM, Emsley HCA, Handley J, Lohezic M, Parkes LM. Compensatory increase in oxygen extraction fraction is associated with age-related cerebrovascular disease. Neuroimage Clin 2025; 45:103746. [PMID: 39922028 DOI: 10.1016/j.nicl.2025.103746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/19/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
Cerebrovascular disease is an important contributor to dementia with reductions in cerebral blood flow (CBF) potentially compromising oxygen supply. In early stages, reduced CBF may be associated with a compensatory increase in oxygen extraction fraction (OEF) to maintain the cerebral metabolic rate of oxygen consumption (CMRO2). We used a simultaneous PET-MRI protocol to measure OEF, CBF, CMRO2, and arterial transit time (ATT) in elderly people (n = 24, age 69.6 ± 5.3 years) with a range of vascular disease risk (QRisk 18.7 ± 10.8 %) and cognitive abilities (MoCA scores 26.7 ± 3.4) to determine if a) vascular disease risk (parameterised with QRisk2 score) is associated with altered CBF, ATT, OEF and CMRO2, b) if impaired blood supply and increasing transit times are associated with elevated OEF and c) if these physiological measures are associated with impaired cognition. ATT rose by 132 ms per 10 point increase in QRisk and there was a trend for reduced CBF. Compensatory increases in OEF occurred in association with modified ATT and CBF, preserving CMRO2. There was no regional variation to these relationships. Cognitive impairment was associated with prolonged ATT. These findings demonstrate the potential use of multi-delay time ASL and Quantitative Susceptibility Mapping for the early detection of cerebrovascular changes and provide evidence for compensatory increases in oxygen extraction in the presence of reduced blood flow.
Collapse
Affiliation(s)
- John McFadden
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Julian Matthews
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Lauren Scott
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Karl Herholz
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Ben Dickie
- Division of Informatics, Imaging and Data Science, School of Health Sciences Manchester UK
| | - Hamied Haroon
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Oliver Sparasci
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Saadat Ahmed
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK
| | - Natalia Kyrtata
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK; University Hospital of Morecambe Bay NHS Foundation Trust Lancaster UK
| | - Geoffrey J M Parker
- Bioxydyn Limited Manchester UK; Centre for Medical Image Computing Department of Medical Physics & Biomedical Engineering and Department of Neuroinflammation University College London London UK
| | - Hedley C A Emsley
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK; Lancaster Medical School Lancaster University Lancaster UK; Department of Neurology Lancashire Teaching Hospitals NHS Foundation Trust Preston UK
| | - Joel Handley
- Department of Neurology Lancashire Teaching Hospitals NHS Foundation Trust Preston UK
| | | | - Laura M Parkes
- Division of Psychology, Communication and Human Neuroscience, School of Health Sciences Faculty of Biology, Medicine, and Health the University of Manchester UK; Geoffrey Jefferson Brain Research Centre Manchester Academic Health Science Centre Manchester UK.
| |
Collapse
|
2
|
Le S, Xu F, Luo Z, Shi W, Lu S, Zhang Z, Guo Z, Xu W, Yang M, Li T, Li X, Liang K, Zhu L. Integrated analysis of chromatin and transcriptomic profiling of the striatum after cerebral hypoperfusion in mice. BMC Genomics 2025; 26:71. [PMID: 39856551 PMCID: PMC11762485 DOI: 10.1186/s12864-025-11256-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Vascular cognitive impairment (VCI) is a significant contributor to dementia, yet the precise mechanisms underlying the cognitive decline associated with chronic cerebral hypoperfusion (CCH) remain unclear. This study investigated the molecular and epigenetic changes in the striatum, a brain region critical for motor function and cognition, following chronic hypoperfusion using a bilateral common carotid artery stenosis (BCAS) model in mice. METHODS RNA-seq was utilized to identify differentially expressed genes (DEGs) associated with hypoperfusion. In parallel, ATAC-seq was used to assess changes in chromatin accessibility within the striatum, providing insight into the epigenome and potential regulatory mechanisms. The integration of these datasets allowed us to correlate chromatin accessibility with transcriptional activity and to identify key transcription factors driving the observed gene expression changes. RESULTS Analysis of striatum-specific transcriptome revealed significant upregulation of immune response genes, particularly type II interferon signaling, and downregulation of neural activation pathways. Analysis of striatum-specific epigenome showed increased chromatin accessibility at promoters of immune-related genes. Integrated analysis highlighted PU.1 as a key transcription factor in upregulated pathways, while neural pathways lacked epigenetic regulation, revealing distinct molecular responses in the striatum following chronic hypoperfusion. CONCLUSIONS Our findings indicate that upregulated pathways in the striatum following BCAS-induced CCH are driven by epigenetic changes, while downregulated pathways occur independently of these modifications. Additionally, PU.1 plays a critical role in mediating immune responses, offering a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Shijia Le
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fengyiyang Xu
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi Luo
- Department of Surgery, Shanghai Deji Hospital, Qingdao University, Shanghai, 200331, China
| | - Weihao Shi
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shuangshuang Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zengyu Zhang
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Zimin Guo
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Wenshi Xu
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Mingqi Yang
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tianyi Li
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Zhongshan Hospital, Institute of Vascular Surgery, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China.
| | - Kun Liang
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Lei Zhu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
3
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Ghaderi S, Mohammadi S, Fatehi F. Current evidence of arterial spin labeling in amyotrophic lateral sclerosis: A systematic review. Clin Neurol Neurosurg 2024; 249:108691. [PMID: 39700696 DOI: 10.1016/j.clineuro.2024.108691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/04/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVE This study aimed to evaluate the utility of arterial spin labeling (ASL) in assessing cerebral blood flow (CBF) changes in amyotrophic lateral sclerosis (ALS), and its potential as a biomarker for early diagnosis. METHODS A systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Studies that employed ASL to compare CBF between ALS patients and healthy controls were included. RESULTS Seven studies were included. A consistent finding across these studies was hypoperfusion in both the motor and non-motor regions, particularly in the frontotemporal cortex. Hypoperfusion in motor regions was correlated with functional impairment and was observed prior to structural changes, suggesting its potential as an early biomarker. There is limited evidence to suggest that monitoring changes in CBF patterns in the brain. Besides, limited findings showed initial hyperperfusion in regions not yet involved in the pathological process, and progressing hypoperfusion in regions with increasing pathological burden. CONCLUSIONS This review highlights the potential of ASL as a valuable tool for understanding the neurovascular dysfunction in ALS. Further research is required to validate its clinical utility for diagnosing ALS and monitoring disease progression.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Neurology, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
5
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
6
|
Chen Z, Liu W, Balu N, Chen L, Ortega D, Huang X, Hatsukami TS, Yang J, Yuan C. Associations of Intracranial Artery Length and Branch Number on Time-of-Flight MRA With Cognitive Impairment in Hypertensive Older Males. J Magn Reson Imaging 2024; 60:1720-1728. [PMID: 38263621 DOI: 10.1002/jmri.29242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Hypertension-induced impairment of the cerebral artery network contributes to cognitive impairment. Characterizing the structure and function of cerebral arteries may facilitate the understanding of hypertension-related pathological mechanisms and lead to the development of new indicators for cognitive impairment. PURPOSE To investigate the associations between morphological features of the intracranial arteries distal to the circle of Willis on time-of-flight MRA (TOF-MRA) and cognitive performance in a hypertensive cohort. STUDY TYPE Prospective observational study. POPULATION 189 hypertensive older males (mean age 64.9 ± 7.2 years). FIELD STRENGTH/SEQUENCE TOF-MRA sequence with a 3D spoiled gradient echo readout and arterial spin labeling perfusion imaging sequence with a 3D stack-of-spirals fast spin echo readout at 3T. ASSESSMENT The intracranial arteries were segmented from TOF-MRA and the total length of distal arteries (TLoDA) and number of arterial branches (NoB) were calculated. The mean gray matter cerebral blood flow (GM-CBF) was extracted from arterial spin labeling perfusion imaging. The cognitive level was assessed with short-term and long-term delay-recall auditory verbal learning test (AVLT) scores, and with montreal cognitive assessment. STATISTICAL TESTS Univariable and multivariable linear regression were used to analyze the associations between TLoDA, NoB, GM-CBF and the cognitive assessment scores, with P < 0.05 indicating significance. RESULTS TLoDA (r = 0.314) and NoB (r = 0.346) were significantly correlated with GM-CBF. Multivariable linear regression analyses showed that TLoDA and NoB, but not GM-CBF (P = 0.272 and 0.141), were significantly associated with short-term and long-term delay-recall AVLT scores. These associations remained significant after adjusting for GM-CBF. DATA CONCLUSION The TLoDA and NoB of distal intracranial arteries on TOF-MRA are significantly associated with cognitive impairment in hypertensive subjects. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Zhensen Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Wenjin Liu
- Department of Nephrology, Clinical Medical College, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, China
- Yangzhou Institute of Precision Medicine for Kidney Diseases, Yangzhou, China
| | - Niranjan Balu
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Li Chen
- Department of Electrical and Computer Engineering, University of Washington, Seattle, Washington, USA
| | - Dakota Ortega
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Xiaoqin Huang
- Department of Nephrology, The First People's Hospital of Yancheng, Yancheng, China
| | - Thomas S Hatsukami
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Junwei Yang
- Center for Kidney Disease, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun Yuan
- Vascular Imaging Lab, Department of Radiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Ölmestig J, Mortensen KN, Fagerlund B, Naveed N, Nordling MM, Christensen H, Iversen HK, Poulsen MB, Siebner HR, Kruuse C. Cerebral blood flow and cognition after 3 months tadalafil treatment in small vessel disease (ETLAS-2): study protocol for a randomized controlled trial. Trials 2024; 25:570. [PMID: 39210472 PMCID: PMC11360322 DOI: 10.1186/s13063-024-08402-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Targeted treatment is highly warranted for cerebral small vessel disease, a causal factor of one in four strokes and a major contributor to vascular dementia. Patients with cerebral small vessel disease have impaired cerebral blood flow and vessel reactivity. Tadalafil is a specific phosphodiesterase 5 inhibitor shown to improve vascular reactivity in the brain. METHODS The ETLAS-2 trial is a phase 2 double-blind, randomized placebo-controlled, parallel trial with the feasibility of tadalafil as the primary outcome. The trial aims to include 100 patients with small vessel occlusion stroke or transitory ischemic attacks and signs of cerebral small vessel disease more than 6 months before administration of study medication. Patients are treated for 3 months with tadalafil 20 mg or placebo daily and undergo magnetic resonance imaging (MRI) to evaluate changes in small vessel disease according to the STandards for ReportIng Vascular changes on nEuroimaging (STRIVE) criteria as well as cerebral blood flow, cerebrovascular reactivity, and neurovascular coupling in a functional MRI sub-study. The investigation includes comprehensive cognitive testing using paper-pencil tests and Cambridge Neuropsychological Test Automated Battery (CANTAB) tests in a cognitive sub-study. DISCUSSION The ETLAS-2 trial tests the feasibility of long-term treatment with tadalafil and explores vascular and cognitive effects in cerebral small vessel disease in trial sub-studies. The study aims to propose a new treatment target and improve the understanding of small vessel disease. Currently, 64 patients have been included and the trial is estimated to be completed in the year 2024. TRIAL REGISTRATION Clinicaltrials.gov, NCT05173896. Registered on 30 December 2021.
Collapse
Affiliation(s)
- Joakim Ölmestig
- Neurovascular Research Unit, Department of Neurology, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, Copenhagen, Denmark
| | - Kristian Nygaard Mortensen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, Copenhagen, Denmark
| | - Birgitte Fagerlund
- Child and Adolescent Mental Health Center, Copenhagen University Hospital, Mental Health Services CPH, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Nadia Naveed
- Department of Radiology, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
| | - Mette Maria Nordling
- Department of Radiology, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark
| | - Hanne Christensen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Helle Klingenberg Iversen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Mai Bang Poulsen
- Department of Neurology, Copenhagen University Hospital-North Zealand, Copenhagen, Denmark
| | - Hartwig Roman Siebner
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital-Amager and Hvidovre, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christina Kruuse
- Neurovascular Research Unit, Department of Neurology, Copenhagen University Hospital-Herlev and Gentofte, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
- Department of Brain and Spinal Cord Injury, Neuroscience Centre, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
8
|
Xu LL, Yang S, Zhou LQ, Chu YH, Pang XW, You YF, Zhang H, Zhang LY, Zhu LF, Chen L, Shang K, Xiao J, Wang W, Tian DS, Qin C. Bruton's tyrosine kinase inhibition ameliorated neuroinflammation during chronic white matter ischemia. J Neuroinflammation 2024; 21:195. [PMID: 39097747 PMCID: PMC11297596 DOI: 10.1186/s12974-024-03187-4] [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: 02/08/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024] Open
Abstract
Chronic cerebral hypoperfusion (CCH), a disease afflicting numerous individuals worldwide, is a primary cause of cognitive deficits, the pathogenesis of which remains poorly understood. Bruton's tyrosine kinase inhibition (BTKi) is considered a promising strategy to regulate inflammatory responses within the brain, a crucial process that is assumed to drive ischemic demyelination progression. However, the potential role of BTKi in CCH has not been investigated so far. In the present study, we elucidated potential therapeutic roles of BTK in both in vitro hypoxia and in vivo ischemic demyelination model. We found that cerebral hypoperfusion induced white matter injury, cognitive impairments, microglial BTK activation, along with a series of microglia responses associated with inflammation, oxidative stress, mitochondrial dysfunction, and ferroptosis. Tolebrutinib treatment suppressed both the activation of microglia and microglial BTK expression. Meanwhile, microglia-related inflammation and ferroptosis processes were attenuated evidently, contributing to lower levels of disease severity. Taken together, BTKi ameliorated white matter injury and cognitive impairments induced by CCH, possibly via skewing microglia polarization towards anti-inflammatory and homeostatic phenotypes, as well as decreasing microglial oxidative stress damage and ferroptosis, which exhibits promising therapeutic potential in chronic cerebral hypoperfusion-induced demyelination.
Collapse
Affiliation(s)
- Lu-Lu Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Vascular Aging, Tongji Hospital of Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Kalantari S, Soltani M, Maghbooli M, Khoshe Mehr F, Kalantari Z, Borji S, Memari B, Hossein Heydari A, Elahi R, Bayat M, Salighehrad H. Alteraciones del flujo sanguíneo cerebral medidas con RM-ASL como predictor de demencia vascular en la enfermedad isquémica de pequeño vaso. RADIOLOGIA 2024. [DOI: 10.1016/j.rx.2024.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Rajeev V, Tabassum NI, Fann DY, Chen CP, Lai MK, Arumugam TV. Intermittent Metabolic Switching and Vascular Cognitive Impairment. J Obes Metab Syndr 2024; 33:92-107. [PMID: 38736362 PMCID: PMC11224924 DOI: 10.7570/jomes24010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/25/2024] [Accepted: 05/08/2024] [Indexed: 05/14/2024] Open
Abstract
Intermittent fasting (IF), a dietary pattern alternating between eating and fasting periods within a 24-hour cycle, has garnered recognition for its potential to enhance both healthspan and lifespan in animal models and humans. It also shows promise in alleviating age-related diseases, including neurodegeneration. Vascular cognitive impairment (VCI) spans a severity range from mild cognitive deficits to severe cognitive deficits and loss of function in vascular dementia. Chronic cerebral hypoperfusion has emerged as a significant contributor to VCI, instigating vascular pathologies such as microbleeds, blood-brain barrier dysfunction, neuronal loss, and white matter lesions. Preclinical studies in rodents strongly suggest that IF has the potential to attenuate pathological mechanisms, including excitotoxicity, oxidative stress, inflammation, and cell death pathways in VCI models. Hence, this supports evaluating IF in clinical trials for both existing and at-risk VCI patients. This review compiles existing data supporting IF's potential in treating VCI-related vascular and neuronal pathologies, emphasizing the mechanisms by which IF may mitigate these issues. Hence providing a comprehensive overview of the available data supporting IF's potential in treating VCI by emphasizing the underlying mechanisms that make IF a promising intervention for VCI.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nishat I. Tabassum
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
| | - David Y. Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Mitchell K.P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Thiruma V. Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Australia
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
11
|
Bohannon DG, Long D, Okhravi HR, Lee SC, De Jesus CL, Neubert TA, Rostagno AA, Ghiso JA, Kim WK. Functionally distinct pericyte subsets differently regulate amyloid-β deposition in patients with Alzheimer's disease. Brain Pathol 2024:e13282. [PMID: 38932696 DOI: 10.1111/bpa.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Although the concept that the blood-brain barrier (BBB) plays an important role in the etiology and pathogenesis of Alzheimer's disease (AD) has become increasingly accepted, little is known yet about how it actually contributes. We and others have recently identified a novel functionally distinct subset of BBB pericytes (PCs). In the present study, we sought to determine whether these PC subsets differentially contribute to AD-associated pathologies by immunohistochemistry and amyloid beta (Aβ) peptidomics. We demonstrated that a disease-associated PC subset (PC2) expanded in AD patients compared to age-matched, cognitively unimpaired controls. Surprisingly, we found that this increase in the percentage of PC2 (%PC2) was correlated negatively with BBB breakdown in AD patients, unlike in natural aging or other reported disease conditions. The higher %PC2 in AD patients was also correlated with a lower Aβ42 plaque load and a lower Aβ42:Aβ40 ratio in the brain as determined by immunohistochemistry. Colocalization analysis of multicolor confocal immunofluorescence microscopy images suggests that AD patient with low %PC2 have higher BBB breakdown due to internalization of Aβ42 by the physiologically normal PC subset (PC1) and their concomitant cell death leading to more vessels without PCs and increased plaque load. On the contrary, it appears that PC2 can secrete cathepsin D to cleave and degrade Aβ built up outside of PC2 into more soluble forms, ultimately contributing to less BBB breakdown and reducing Aβ plaque load. Collectively our data shows functionally distinct mechanisms for PC1 and PC2 in high Aβ conditions, demonstrating the importance of correctly identifying these populations when investigating the contribution of neurovascular dysfunction to AD pathogenesis.
Collapse
Affiliation(s)
- Diana G Bohannon
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Danielle Long
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Hamid R Okhravi
- Glennan Center for Geriatrics and Gerontology, Eastern Virginia Medical School, Norfolk, Virginia, USA
- Integrated Neurodegenerative Disorders Center, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Sunhee C Lee
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | | | - Thomas A Neubert
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Agueda A Rostagno
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Jorge A Ghiso
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
- Integrated Neurodegenerative Disorders Center, Eastern Virginia Medical School, Norfolk, Virginia, USA
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
12
|
Ugodnikov A, Persson H, Simmons CA. Bridging barriers: advances and challenges in modeling biological barriers and measuring barrier integrity in organ-on-chip systems. LAB ON A CHIP 2024; 24:3199-3225. [PMID: 38689569 DOI: 10.1039/d3lc01027a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Biological barriers such as the blood-brain barrier, skin, and intestinal mucosal barrier play key roles in homeostasis, disease physiology, and drug delivery - as such, it is important to create representative in vitro models to improve understanding of barrier biology and serve as tools for therapeutic development. Microfluidic cell culture and organ-on-a-chip (OOC) systems enable barrier modelling with greater physiological fidelity than conventional platforms by mimicking key environmental aspects such as fluid shear, accurate microscale dimensions, mechanical cues, extracellular matrix, and geometrically defined co-culture. As the prevalence of barrier-on-chip models increases, so does the importance of tools that can accurately assess barrier integrity and function without disturbing the carefully engineered microenvironment. In this review, we first provide a background on biological barriers and the physiological features that are emulated through in vitro barrier models. Then, we outline molecular permeability and electrical sensing barrier integrity assessment methods, and the related challenges specific to barrier-on-chip implementation. Finally, we discuss future directions in the field, as well important priorities to consider such as fabrication costs, standardization, and bridging gaps between disciplines and stakeholders.
Collapse
Affiliation(s)
- Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Henrik Persson
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
13
|
Hu Z, Li Z, Shi Y, Liu S, Shen Y, Hu F, Li Q, Liu X, Gou X, Chen Z, Yang D. Advancements in investigating the role of cerebral small vein loss in Alzheimer's disease-related pathological changes. Neurol Sci 2024; 45:1875-1883. [PMID: 38133856 DOI: 10.1007/s10072-023-07208-7] [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: 08/16/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023]
Abstract
Alzheimer's disease (AD) is the prevailing type of dementia in the elderly, yet a comprehensive comprehension of its precise underlying mechanisms remains elusive. The investigation of the involvement of cerebral small veins in the advancement of AD has yet to be sufficiently explored in previous studies, primarily due to constraints associated with pathological staining techniques. However, recent research has provided valuable insights into multiple pathophysiological occurrences concerning cerebral small veins in AD, which may manifest sequentially, concurrently, or in a self-perpetuating manner. These events are presumed to be among the initial processes in the disease's progression. The impact of cerebral small vein loss on amyloid beta (Aβ) clearance through the glial lymphatic system is noteworthy. There exists a potential interdependence between collagen deposition and Aβ deposition in cerebral small veins. The compromised functionality of cerebral small veins can result in decreased cerebral perfusion pressure, potentially leading to cerebral tissue ischemia and edema. Additionally, the reduction of cerebral small veins may facilitate the infiltration of inflammatory factors into the brain parenchyma, thereby eliciting neuroinflammatory responses. Susceptibility-weighted imaging (SWI) is a valuable modality for the efficient assessment of cerebral small veins, precisely the deep medullary vein (DMV), and holds promise for the identification of precise and reliable imaging biomarkers for AD. This review presents a comprehensive overview of the current advancements and obstacles to the impairment of cerebral small veins in AD. Additionally, we emphasize future research avenues and the importance of conducting further investigations in this domain.
Collapse
Affiliation(s)
- Zhenzhu Hu
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Zhaoying Li
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Yu Shi
- Department of Neurology, Xuzhou Cancer Hospital, Xuzhou, 221000, Jiangsu, China
| | - Shanyu Liu
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Yuling Shen
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Fangfang Hu
- Department of Neurology, Xuzhou Cancer Hospital, Xuzhou, 221000, Jiangsu, China
| | - Qingqing Li
- Department of Neurology, Xuzhou Cancer Hospital, Xuzhou, 221000, Jiangsu, China
| | - Xu Liu
- Department of Neurology, Xuzhou Cancer Hospital, Xuzhou, 221000, Jiangsu, China
| | - Xinyu Gou
- Department of Neurology, Guang'an People's Hospital, Guang'an, 638001, China
| | - Zhenwei Chen
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China
| | - Dongdong Yang
- Department of Neurology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610032, China.
| |
Collapse
|
14
|
Ni YC, Lin ZK, Cheng CH, Pai MC, Chiu PY, Chang CC, Chang YT, Hung GU, Lin KJ, Hsiao IT, Lin CY, Yang HC. Classification Prediction of Alzheimer's Disease and Vascular Dementia Using Physiological Data and ECD SPECT Images. Diagnostics (Basel) 2024; 14:365. [PMID: 38396404 PMCID: PMC10888136 DOI: 10.3390/diagnostics14040365] [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: 12/06/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are the two most common forms of dementia. However, their neuropsychological and pathological features often overlap, making it difficult to distinguish between AD and VaD. In addition to clinical consultation and laboratory examinations, clinical dementia diagnosis in Taiwan will also include Tc-99m-ECD SPECT imaging examination. Through machine learning and deep learning technology, we explored the feasibility of using the above clinical practice data to distinguish AD and VaD. We used the physiological data (33 features) and Tc-99m-ECD SPECT images of 112 AD patients and 85 VaD patients in the Taiwanese Nuclear Medicine Brain Image Database to train the classification model. The results, after filtering by the number of SVM RFE 5-fold features, show that the average accuracy of physiological data in distinguishing AD/VaD is 81.22% and the AUC is 0.836; the average accuracy of training images using the Inception V3 model is 85% and the AUC is 0.95. Finally, Grad-CAM heatmap was used to visualize the areas of concern of the model and compared with the SPM analysis method to further understand the differences. This research method can quickly use machine learning and deep learning models to automatically extract image features based on a small amount of general clinical data to objectively distinguish AD and VaD.
Collapse
Affiliation(s)
- Yu-Ching Ni
- Department of Radiation Protection, National Atomic Research Institute, Taoyuan 325, Taiwan
| | - Zhi-Kun Lin
- Department of Radiation Protection, National Atomic Research Institute, Taoyuan 325, Taiwan
| | - Chen-Han Cheng
- Department of Radiation Protection, National Atomic Research Institute, Taoyuan 325, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Institute of Gerontology, National Cheng Kung University, Tainan 701, Taiwan
- Alzheimer’s Disease Research Center, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Pai-Yi Chiu
- Department of Neurology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Institute of Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Ya-Ting Chang
- Department of Neurology, Institute of Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Guang-Uei Hung
- Department of Nuclear Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 505, Taiwan
| | - Kun-Ju Lin
- Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Molecular Imaging Center and Department of Nuclear Medicine, Chang Gung University and Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ing-Tsung Hsiao
- Healthy Aging Research Center and Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Molecular Imaging Center and Department of Nuclear Medicine, Chang Gung University and Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chia-Yu Lin
- Department of Radiation Protection, National Atomic Research Institute, Taoyuan 325, Taiwan
| | - Hui-Chieh Yang
- Department of Radiation Protection, National Atomic Research Institute, Taoyuan 325, Taiwan
| |
Collapse
|
15
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
16
|
van Dinther M, Hooghiemstra AM, Bron EE, Versteeg A, Leeuwis AE, Kalay T, Moonen JE, Kuipers S, Backes WH, Jansen JFA, van Osch MJP, Biessels G, Staals J, van Oostenbrugge RJ. Lower cerebral blood flow predicts cognitive decline in patients with vascular cognitive impairment. Alzheimers Dement 2024; 20:136-144. [PMID: 37491840 PMCID: PMC10917014 DOI: 10.1002/alz.13408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Chronic cerebral hypoperfusion is one of the assumed pathophysiological mechanisms underlying vascular cognitive impairment (VCI). We investigated the association between baseline cerebral blood flow (CBF) and cognitive decline after 2 years in patients with VCI and reference participants. METHODS One hundred eighty-one participants (mean age 66.3 ± 7.4 years, 43.6% women) underwent arterial spin labeling (ASL) magnetic resonance imaging (MRI) and neuropsychological assessment at baseline and at 2-year follow-up. We determined the association between baseline global and lobar CBF and cognitive decline with multivariable regression analysis. RESULTS Lower global CBF at baseline was associated with more global cognitive decline in VCI and reference participants. This association was most profound in the domain of attention/psychomotor speed. Lower temporal and frontal CBF at baseline were associated with more cognitive decline in memory. DISCUSSION Our study supports the role of hypoperfusion in the pathophysiological and clinical progression of VCI. HIGHLIGHTS Impaired cerebral blood flow (CBF) at baseline is associated with faster cognitive decline in VCI and normal aging. Our results suggest that low CBF precedes and contributes to the development of vascular cognitive impairment. CBF determined by ASL might be used as a biomarker to monitor disease progression or treatment responses in VCI.
Collapse
Affiliation(s)
- Maud van Dinther
- Department of NeurologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Astrid M. Hooghiemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Esther E. Bron
- Department of Radiology & Nuclear MedicineErasmus MC—University Medical Center RotterdamRotterdamThe Netherlands
| | - Adriaan Versteeg
- Department of Radiology & Nuclear MedicineErasmus MC—University Medical Center RotterdamRotterdamThe Netherlands
| | - Anna E. Leeuwis
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Old Age PsychiatryGGZ inGeestAmsterdamThe Netherlands
| | - Tugba Kalay
- Department of NeurologySt. Antonius ZiekenhuisNieuwegeinThe Netherlands
| | - Justine E. Moonen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sanne Kuipers
- Department of NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Walter H. Backes
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Jacobus F. A. Jansen
- Department of Radiology and Nuclear MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Mathias J. P. van Osch
- C.J. Gorter MRI Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Geert‐Jan Biessels
- Department of NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Julie Staals
- Department of NeurologyMaastricht University Medical CenterMaastrichtThe Netherlands
| | | | | |
Collapse
|
17
|
Kandiah N. Cognitive Outcomes Poststroke: A Need for Better Insights into Mechanisms. Brain Connect 2023; 13:438-440. [PMID: 37782227 DOI: 10.1089/brain.2023.29054.editorial] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Affiliation(s)
- Nagaendran Kandiah
- Associate Professor of Neuroscience and Mental Health, Nanyang Technological University, Singapore, Singapore
- Director, Dementia Research Centre (Singapore), LKC-Imperial Medical School, Nanyang Technological University, Singapore, Singapore
- Consultant Neurologist, National University Hospital, Singapore, Singapore
- Clinician Scientist, National Medical Research Council, Singapore, Singapore
| |
Collapse
|
18
|
Badji A, Youwakim J, Cooper A, Westman E, Marseglia A. Vascular cognitive impairment - Past, present, and future challenges. Ageing Res Rev 2023; 90:102042. [PMID: 37634888 DOI: 10.1016/j.arr.2023.102042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Vascular cognitive impairment (VCI) is a lifelong process encompassing a broad spectrum of cognitive disorders, ranging from subtle or mild deficits to prodromal and fully developed dementia, originating from cerebrovascular lesions such as large and small vessel disease. Genetic predisposition and environmental exposure to risk factors such as unhealthy lifestyles, hypertension, cardiovascular disease, and metabolic disorders will synergistically interact, yielding biochemical and structural brain changes, ultimately culminating in VCI. However, little is known about the pathological processes underlying VCI and the temporal dynamics between risk factors and disease mechanisms (biochemical and structural brain changes). This narrative review aims to provide an evidence-based summary of the link between individual vascular risk/disorders and cognitive dysfunction and the potential structural and biochemical pathophysiological processes. We also discuss some key challenges for future research on VCI. There is a need to shift from individual risk factors/disorders to comorbid vascular burden, identifying and integrating imaging and fluid biomarkers, implementing a life-course approach, considering possible neuroprotective influences of positive life exposures, and addressing biological sex at birth and gender differences. Finally, this review highlights the need for future researchers to leverage and integrate multidimensional data to advance our understanding of the mechanisms and pathophysiology of VCI.
Collapse
Affiliation(s)
- Atef Badji
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Youwakim
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage (CIRCA), Montreal, QC, Canada; Groupe de Recherche sur la Signalisation Neuronal et la Circuiterie (SNC), Montreal, QC, Canada
| | - Alexandra Cooper
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Anna Marseglia
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
20
|
Swinford CG, Risacher SL, Wu YC, Apostolova LG, Gao S, Bice PJ, Saykin AJ. Altered cerebral blood flow in older adults with Alzheimer's disease: a systematic review. Brain Imaging Behav 2023; 17:223-256. [PMID: 36484922 PMCID: PMC10117447 DOI: 10.1007/s11682-022-00750-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/26/2022] [Accepted: 11/20/2022] [Indexed: 12/13/2022]
Abstract
The prevalence of Alzheimer's disease is projected to reach 13 million in the U.S. by 2050. Although major efforts have been made to avoid this outcome, so far there are no treatments that can stop or reverse the progressive cognitive decline that defines Alzheimer's disease. The utilization of preventative treatment before significant cognitive decline has occurred may ultimately be the solution, necessitating a reliable biomarker of preclinical/prodromal disease stages to determine which older adults are most at risk. Quantitative cerebral blood flow is a promising potential early biomarker for Alzheimer's disease, but the spatiotemporal patterns of altered cerebral blood flow in Alzheimer's disease are not fully understood. The current systematic review compiles the findings of 81 original studies that compared resting gray matter cerebral blood flow in older adults with mild cognitive impairment or Alzheimer's disease and that of cognitively normal older adults and/or assessed the relationship between cerebral blood flow and objective cognitive function. Individuals with Alzheimer's disease had relatively decreased cerebral blood flow in all brain regions investigated, especially the temporoparietal and posterior cingulate, while individuals with mild cognitive impairment had consistent results of decreased cerebral blood flow in the posterior cingulate but more mixed results in other regions, especially the frontal lobe. Most papers reported a positive correlation between regional cerebral blood flow and cognitive function. This review highlights the need for more studies assessing cerebral blood flow changes both spatially and temporally over the course of Alzheimer's disease, as well as the importance of including potential confounding factors in these analyses.
Collapse
Affiliation(s)
- Cecily G Swinford
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Paula J Bice
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th St. IU Neuroscience Center, GH 4101, 46202, Indianapolis, IN, USA.
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
21
|
Hu Y, Zhang M, Liu B, Tang Y, Wang Z, Wang T, Zheng J, Zhang J. Honokiol prevents chronic cerebral hypoperfusion induced astrocyte A1 polarization to alleviate neurotoxicity by targeting SIRT3-STAT3 axis. Free Radic Biol Med 2023; 202:62-75. [PMID: 36997099 DOI: 10.1016/j.freeradbiomed.2023.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
Alzheimer's Dementia (AD) and Vascular Dementia (VaD) are two main types of dementias for which no specific treatment is available. Chronic Cerebral Hypoperfusion (CCH) is a pathogenesis underlying AD and VaD that promotes neuroinflammatory responses and oxidative stress. Honokiol (HNK) is a natural compound isolated from magnolia leaves that can easily cross blood brain barrier and has anti-inflammatory and antioxidant effects. In the present study, the effects of HNK on astrocyte polarization and neurological damage in in vivo and in vitro models of chronic cerebral hypoperfusion were explored. We found that HNK was able to inhibit the phosphorylation and nuclear translocation of STAT3, A1 polarization, and reduce conditioned medium's neuronal toxicity of astrocyte under chronic hypoxia induced by cobalt chloride; STAT3 phosphorylation inhibitor C188-9 was able to mimic the above effects of HNK, suggesting that HNK may inhibit chronic hypoxia-induced A1 polarization in astrocytes via STAT3. SIRT3 inhibitor 3-TYP reversed, while Sirt3 overexpression mimicked the inhibitory effects of HNK on oxidative stress, STAT3 phosphorylation and nuclear translocation, A1 polarization and neuronal toxicity of astrocyte under chronic hypoxic conditions. For in vivo research, continuous intraperitoneal injection of HNK (1mg/kg) for 21 days ameliorated the decrease in SIRT3 activity and oxidative stress, inhibited astrocytic STAT3 nuclear translocation and A1 polarization, and prevented neuron and synaptic loss in the hippocampal of CCH rats. Besides, HNK application improved the spatial memory impairment of CCH rats, as assessed with Morris Water Maze. In conclusion, these results suggest that the phytochemical HNK can inhibit astrocyte A1 polarization via regulating SIRT3-STAT3 axis, thus improving CCH-induced neurological damage. These results highlight HNK as novel treatment for dementia with underlying vascular mechanisms.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Bihan Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Yingying Tang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Tao Wang
- Department of Neurology, First Clinical Medical College of China Three Gorges University, Yichang, Hubei, 443003, China
| | - Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| |
Collapse
|
22
|
Liu Q, Zhang X. Multimodality neuroimaging in vascular mild cognitive impairment: A narrative review of current evidence. Front Aging Neurosci 2023; 15:1073039. [PMID: 37009448 PMCID: PMC10050753 DOI: 10.3389/fnagi.2023.1073039] [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: 10/18/2022] [Accepted: 02/24/2023] [Indexed: 03/17/2023] Open
Abstract
The vascular mild cognitive impairment (VaMCI) is generally accepted as the premonition stage of vascular dementia (VaD). However, most studies are focused mainly on VaD as a diagnosis in patients, thus neglecting the VaMCI stage. VaMCI stage, though, is easily diagnosed by vascular injuries and represents a high-risk period for the future decline of patients' cognitive functions. The existing studies in China and abroad have found that magnetic resonance imaging technology can provide imaging markers related to the occurrence and development of VaMCI, which is an important tool for detecting the changes in microstructure and function of VaMCI patients. Nevertheless, most of the existing studies evaluate the information of a single modal image. Due to the different imaging principles, the data provided by a single modal image are limited. In contrast, multi-modal magnetic resonance imaging research can provide multiple comprehensive data such as tissue anatomy and function. Here, a narrative review of published articles on multimodality neuroimaging in VaMCI diagnosis was conducted,and the utilization of certain neuroimaging bio-markers in clinical applications was narrated. These markers include evaluation of vascular dysfunction before tissue damages and quantification of the extent of network connectivity disruption. We further provide recommendations for early detection, progress, prompt treatment response of VaMCI, as well as optimization of the personalized treatment plan.
Collapse
Affiliation(s)
- Qiuping Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
23
|
Hnilicova P, Kantorova E, Sutovsky S, Grofik M, Zelenak K, Kurca E, Zilka N, Parvanovova P, Kolisek M. Imaging Methods Applicable in the Diagnostics of Alzheimer's Disease, Considering the Involvement of Insulin Resistance. Int J Mol Sci 2023; 24:3325. [PMID: 36834741 PMCID: PMC9958721 DOI: 10.3390/ijms24043325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease and the most frequently diagnosed type of dementia, characterized by (1) perturbed cerebral perfusion, vasculature, and cortical metabolism; (2) induced proinflammatory processes; and (3) the aggregation of amyloid beta and hyperphosphorylated Tau proteins. Subclinical AD changes are commonly detectable by using radiological and nuclear neuroimaging methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and single-photon emission computed tomography (SPECT). Furthermore, other valuable modalities exist (in particular, structural volumetric, diffusion, perfusion, functional, and metabolic magnetic resonance methods) that can advance the diagnostic algorithm of AD and our understanding of its pathogenesis. Recently, new insights into AD pathoetiology revealed that deranged insulin homeostasis in the brain may play a role in the onset and progression of the disease. AD-related brain insulin resistance is closely linked to systemic insulin homeostasis disorders caused by pancreas and/or liver dysfunction. Indeed, in recent studies, linkages between the development and onset of AD and the liver and/or pancreas have been established. Aside from standard radiological and nuclear neuroimaging methods and clinically fewer common methods of magnetic resonance, this article also discusses the use of new suggestive non-neuronal imaging modalities to assess AD-associated structural changes in the liver and pancreas. Studying these changes might be of great clinical importance because of their possible involvement in AD pathogenesis during the prodromal phase of the disease.
Collapse
Affiliation(s)
- Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ema Kantorova
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 813 67 Bratislava, Slovakia
| | - Milan Grofik
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Kamil Zelenak
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Egon Kurca
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
24
|
Bah TM, Siler DA, Ibrahim AH, Cetas JS, Alkayed NJ. Fluid dynamics in aging-related dementias. Neurobiol Dis 2023; 177:105986. [PMID: 36603747 DOI: 10.1016/j.nbd.2022.105986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/03/2023] Open
Abstract
Recent human and animal model experimental studies revealed novel pathways for fluid movement, immune cell trafficking and metabolic waste clearance in CNS. These studies raise the intriguing possibility that the newly discovered pathways, including the glymphatic system, lymphatic meningeal vessels and skull-brain communication channels, are impaired in aging and neurovascular and neurodegenerative diseases associated with dementia, including Alzheimer's disease (AD) and AD-related dementia. We provide an overview of the glymphatic and dural meningeal lymphatic systems, review current methods and approaches used to study glymphatic flow in humans and animals, and discuss current evidence and controversies related to its role in CNS flow homeostasis under physiological and pathophysiological conditions. Non-invasive imaging approaches are needed to fully understand the mechanisms and pathways driving fluid movement in CNS and their roles across lifespan including healthy aging and aging-related dementia.
Collapse
Affiliation(s)
- Thierno M Bah
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Dominic A Siler
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Aseel H Ibrahim
- Department of Neurosurgery, University of Arizona, Tucson, AZ, USA
| | - Justin S Cetas
- Department of Neurosurgery, University of Arizona, Tucson, AZ, USA
| | - Nabil J Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
25
|
Huang D, Guo Y, Guan X, Pan L, Zhu Z, Chen Z, Dijkhuizen RM, Duering M, Yu F, Boltze J, Li P. Recent advances in arterial spin labeling perfusion MRI in patients with vascular cognitive impairment. J Cereb Blood Flow Metab 2023; 43:173-184. [PMID: 36284489 PMCID: PMC9903225 DOI: 10.1177/0271678x221135353] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/01/2022] [Accepted: 09/21/2022] [Indexed: 01/24/2023]
Abstract
Cognitive impairment (CI) is a major health concern in aging populations. It impairs patients' independent life and may progress to dementia. Vascular cognitive impairment (VCI) encompasses all cerebrovascular pathologies that contribute to cognitive impairment (CI). Moreover, the majority of CI subtypes involve various aspects of vascular dysfunction. Recent research highlights the critical role of reduced cerebral blood flow (CBF) in the progress of VCI, and the detection of altered CBF may help to detect or even predict the onset of VCI. Arterial spin labeling (ASL) is a non-invasive, non-ionizing perfusion MRI technique for assessing CBF qualitatively and quantitatively. Recent methodological advances enabling improved signal-to-noise ratio (SNR) and data acquisition have led to an increase in the use of ASL to assess CBF in VCI patients. Combined with other imaging modalities and biomarkers, ASL has great potential for identifying early VCI and guiding prediction and prevention strategies. This review focuses on recent advances in ASL-based perfusion MRI for identifying patients at high risk of VCI.
Collapse
Affiliation(s)
- Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunlu Guo
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Guan
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijun Pan
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeng’ai Chen
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany
- Medical Image Analysis Center (MIAC) and qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Fang Yu
- Department of Anesthesiology, Westchester Medical Center, New York Medical College, NY, USA
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Lu Z, Teng Y, Wang L, Jiang Y, Li T, Chen S, Wang B, Li Y, Yang J, Wu X, Cheng W, Cui X, Zhao M. Abnormalities of hippocampus and frontal lobes in heart failure patients and animal models with cognitive impairment or depression: A systematic review. PLoS One 2022; 17:e0278398. [PMID: 36490252 PMCID: PMC9733898 DOI: 10.1371/journal.pone.0278398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
AIMS This systematic review aimed to study the hippocampal and frontal changes of heart failure (HF) patients and HF animal models with cognitive impairment or depression. METHODS A systematic review of the literature was conducted independently by reviewers using PubMed, Web of Science, Embase, and the Cochrane Library databases. RESULTS AND CONCLUSIONS 30 studies were included, involving 17 pieces of clinical research on HF patients and 13 studies of HF animal models. In HF patients, the hippocampal injuries were shown in the reduction of volume, CBF, glucose metabolism, and gray matter, which were mainly observed in the right hippocampus. The frontal damages were only in reduced gray matter and have no difference between the right and left sides. The included HF animal model studies were generalized and demonstrated the changes in inflammation and apoptosis, synaptic reduction, and neurotransmitter disorders in the hippocampus and frontal lobes. The results of HF animal model studies complemented the clinical observations by providing potential mechanistic explanations of the changes in the hippocampus and frontal lobes.
Collapse
Affiliation(s)
- Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yangyang Jiang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tong Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shiqi Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingjing Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weiting Cheng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- * E-mail: (MZ); (XC)
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- * E-mail: (MZ); (XC)
| |
Collapse
|
27
|
Chiara C, Gilda C, Daniela M, Antonio C, Miriana M, Marcello M, Elena S, Roberta L, Ciro C, Vincenzo BM. A two-year longitudinal study of retinal vascular impairment in patients with amnestic mild cognitive impairment. Front Aging Neurosci 2022; 14:993621. [PMID: 36420311 PMCID: PMC9678013 DOI: 10.3389/fnagi.2022.993621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
ObjectiveTo evaluate the relation between retinal vascular impairment and cognitive decline in patients with amnestic mild cognitive impairment (aMCI) over time.MethodsSpectral domain-optical coherence tomography (SD-OCT) and OCT angiography study was performed in aMCI patients over 2 years follow-up and compared to baseline.ResultsThirty-eight eyes from 19 aMCI patients were evaluated. Structural and vascular OCT measures were reduced at follow-up except for vessel density (VD) of the choriocapillaris, unchanged, and foveal avascular zone, which was increased; no changes in any parameter were found in 18 age-matched healthy controls. Overall, these findings were confirmed when patients were evaluated separately according to progression to dementia. Only non-converters to dementia showed significant VD reduction in the deep capillary plexuses (coeff. β = −4.20; p < 0.001), may be for an initial massive VD depletion becoming less evident with progression of the disease. MMSE reduction was associated with a higher ganglion cell complex reduction (coeff. β = 0.10; p = 0.04) and a higher VD reduction in the radial peripapillary capillary (RPC) plexus (coeff. β = 0.14; p = 0.02) in the whole patient group, while it was associated with a higher VD reduction only in RPC plexus in converters (coeff. β = 0.21; p < 0.001).ConclusionOur data shows vascular impairment progression in the inner retina of aMCI patients and support the hypothesis that vascular changes may contribute to the onset and progression of Alzheimer’s disease. Other follow-up studies, with a larger number of patients, are needed to better define VD as a potential biomarker.
Collapse
|
28
|
Zhang W, Li M, Zhou X, Huang C, Wan K, Li C, Yin J, Zhao W, Zhang C, Zhu X, Sun Z. Altered serum amyloid beta and cerebral perfusion and their associations with cognitive function in patients with subcortical ischemic vascular disease. Front Neurosci 2022; 16:993767. [PMID: 36312019 PMCID: PMC9608371 DOI: 10.3389/fnins.2022.993767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/08/2022] [Indexed: 01/07/2024] Open
Abstract
Subcortical ischemic vascular disease (SIVD) is one of the important causes of cognitive dysfunction, altered amyloid-beta (Aβ) and cerebral perfusion may be involved in the pathophysiological mechanism of SIVD and are closely related to cognitive function. We aimed to investigate altered serum Aβ and cerebral perfusion in patients with SIVD and their correlation with cognitive function. Seventy-four healthy controls (HCs) and 74 SIVD patients, including 38 SIVD patients with no cognitive impairment (SIVD-NCI) and 36 SIVD patients with mild cognitive impairment (SIVD-MCI) underwent the measurement of serum Aβ40 and Aβ42 levels, pseudo-continuous arterial spin labeling MRI scanning, and cognitive evaluation. Compared to the healthy controls (HCs), the level of serum Aβ40 and Aβ40/42 ratio increased and Aβ42 decreased in SIVD patients. The serum Aβ40 level and Aβ40/42 ratio in patients with SIVD-MCI were significantly higher than those in the HCs and SIVD-NCI, and the level of Aβ42 in the SIVD-MCI was lower than the HCs. In addition, the serum Aβ40/42 ratio provided high diagnostic accuracy for SIVD and SIVD-MCI, it was further identified as an independent risk factor for cognitive impairment. Patients with SIVD-NCI and SIVD-MCI exhibited both increased and decreased cerebral blood flow (CBF) in regional. The Aβ40/42 ratio was associated with global CBF, while altered global and regional CBF was associated with cognitive deficits. In addition, white matter hyperintensities volume (WMHV) correlated with Aβ40/42 ratio, CBF, and cognition. The relationship between Aβ40/42 ratio and cognition was partially mediated by altered CBF. Based on these results, we conclude that the serum Aβ40/42 ratio may be a potential biomarker that can complement current methods for the prediction and diagnosis of cognitive impairment in SIVD patients. In addition, serum Aβ may play a role in cognitive function by regulating CBF, which provides new insights into the intervention, treatment, and prevention of cognitive impairment in SIVD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingxu Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xia Zhou
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chaojuan Huang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ke Wan
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenchen Li
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiabin Yin
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenming Zhao
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cun Zhang
- Department of Radiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoqun Zhu
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongwu Sun
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
29
|
Bhatia K, Kindelin A, Nadeem M, Khan MB, Yin J, Fuentes A, Miller K, Turner GH, Preul MC, Ahmad AS, Mufson EJ, Waters MF, Ahmad S, Ducruet AF. Complement C3a Receptor (C3aR) Mediates Vascular Dysfunction, Hippocampal Pathology, and Cognitive Impairment in a Mouse Model of VCID. Transl Stroke Res 2022; 13:816-829. [PMID: 35258803 DOI: 10.1007/s12975-022-00993-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/12/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) secondary to chronic mild-moderate cerebral ischemia underlie a significant percentage of cases of dementia. We previously reported that either genetic deficiency of the complement C3a receptor (C3aR) or its pharmacological inhibition protects against cerebral ischemia in rodents, while others have implicated C3aR in the pathogenesis seen in rodent transgenic models of Alzheimer's disease. In the present study, we evaluated the role of complement C3a-C3aR signaling in the onset and progression of VCID. We utilized the bilateral common carotid artery stenosis (BCAS) model to induce VCID in male C57BL/6 wild-type and C3aR-knockout (C3aR-/-) mice. Cerebral blood flow (CBF) changes, hippocampal atrophy (HA), white matter degeneration (WMD), and ventricular size were assessed at 4 months post-BCAS using laser speckle contrast analysis (LSCI) and magnetic resonance imaging (MRI). Cognitive function was evaluated using the Morris water maze (MWM), and novel object recognition (NOR), immunostaining, and western blot were performed to assess the effect of genetic C3aR deletion on post-VCID outcomes. BCAS resulted in decreased CBF and increased HA, WMD, and neurovascular inflammation in WT (C57BL/6) compared to C3aR-/- (C3aR-KO) mice. Moreover, C3aR-/- mice exhibited improved cognitive function on NOR and MWM relative to WT controls. We conclude that over-activation of the C3a/C3aR axis exacerbates neurovascular inflammation leading to poor VCID outcomes which are mitigated by C3aR deletion. Future studies are warranted to dissect the role of cell-specific C3aR in VCID.
Collapse
Affiliation(s)
- Kanchan Bhatia
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ, USA
| | - Adam Kindelin
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Muhammad Nadeem
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | | | - Junxiang Yin
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Alberto Fuentes
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Karis Miller
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Gregory H Turner
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Mark C Preul
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Abdullah S Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Michael F Waters
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Saif Ahmad
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
| | - Andrew F Ducruet
- Departments of Neurosurgery & Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85086, USA.
| |
Collapse
|
30
|
Gyanwali B, Tan CS, Petr J, Escobosa LLT, Vrooman H, Chen C, Mutsaerts HJ, Hilal S. Arterial Spin-Labeling Parameters and Their Associations with Risk Factors, Cerebral Small-Vessel Disease, and Etiologic Subtypes of Cognitive Impairment and Dementia. AJNR Am J Neuroradiol 2022; 43:1418-1423. [PMID: 36562454 PMCID: PMC9575536 DOI: 10.3174/ajnr.a7630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/01/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral small-vessel disease may alter cerebral blood flow (CBF) leading to brain changes and, hence, cognitive impairment and dementia. CBF and the spatial coefficient of variation can be measured quantitatively by arterial spin-labeling. We aimed to investigate the associations of demographics, vascular risk factors, location, and severity of cerebral small-vessel disease as well as the etiologic subtypes of cognitive impairment and dementia with CBF and the spatial coefficient of variation. MATERIALS AND METHODS Three hundred ninety patients with a diagnosis of no cognitive impairment, cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia were recruited from the memory clinic. Cerebral microbleeds and lacunes were categorized into strictly lobar, strictly deep, and mixed-location and enlarged perivascular spaces into the centrum semiovale and basal ganglia. Total and region-specific white matter hyperintensity volumes were segmented using FreeSurfer. CBF (n = 333) and the spatial coefficient of variation (n = 390) were analyzed with ExploreASL from 2D-EPI pseudocontinuous arterial spin-labeling images in white matter (WM) and gray matter (GM). To analyze the effect of demographic and vascular risk factors as well as the location and severity of cerebral small-vessel disease markers on arterial spin-labeling parameters, we constructed linear regression models, whereas logistic regression models were used to determine the association between arterial spin-labeling parameters and cognitive impairment no dementia, vascular cognitive impairment no dementia, Alzheimer disease, and vascular dementia. RESULTS Increasing age, male sex, hypertension, hyperlipidemia, history of heart disease, and smoking were associated with lower CBF and a higher spatial coefficient of variation. Higher numbers of lacunes and cerebral microbleeds were associated with lower CBF and a higher spatial coefficient of variation. Location-specific analysis showed mixed-location lacunes and cerebral microbleeds were associated with lower CBF. Higher total, anterior, and posterior white matter hyperintensity volumes were associated with a higher spatial coefficient of variation. No association was observed between enlarged perivascular spaces and arterial spin-labeling parameters. A higher spatial coefficient of variation was associated with the diagnosis of vascular cognitive impairment no dementia, Alzheimer's disease, and vascular dementia. CONCLUSIONS Reduced CBF and an increased spatial coefficient of variation were associated with cerebral small-vessel disease, and more specifically lacunes, whereas cerebral microbleeds and white matter hyperintensities were associated with WM-CBF and GM spatial coefficient of variation. The spatial coefficient of variation was associated with cognitive impairment and dementia, suggesting that hypoperfusion might be the key underlying mechanism for vascular brain damage.
Collapse
Affiliation(s)
- B Gyanwali
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
| | - C S Tan
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - J Petr
- Helmholtz-Zentrum Dresden-Rossendorf (J.P.), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - L L T Escobosa
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
| | - H Vrooman
- Department of Radiology and Nuclear Medicine (H.V.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - C Chen
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - H J Mutsaerts
- Department of Radiology (H.J.M.), VU University Medical Center, Amsterdam, the Netherlands
- Department of Radiology (H.J.M.), Brain Center Rudolf Magnus, University Medical Center, Utrecht, the Netherlands
| | - S Hilal
- From the Memory Aging and Cognition Centre (B.G., C.C., S.H.), National University Health System, Singapore
- Saw Swee Hock School of Public Health (C.S.T., L.L.T.E., S.H.), National University of Singapore, and National University Health System, Singapore
- Department of Pharmacology (C.C., S.H.), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
31
|
Binnie LR, Pauls MMH, Benjamin P, Dhillon MPK, Betteridge S, Clarke B, Ghatala R, Hainsworth FAH, Howe FA, Khan U, Kruuse C, Madigan JB, Moynihan B, Patel B, Pereira AC, Rostrup E, Shtaya ABY, Spilling CA, Trippier S, Williams R, Isaacs JD, Barrick TR, Hainsworth AH. Test-retest reliability of arterial spin labelling for cerebral blood flow in older adults with small vessel disease. Transl Stroke Res 2022; 13:583-594. [PMID: 35080734 PMCID: PMC9232403 DOI: 10.1007/s12975-021-00983-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 12/03/2022]
Abstract
Cerebral small vessel disease (SVD) is common in older people and is associated with lacunar stroke, white matter hyperintensities (WMH) and vascular cognitive impairment. Cerebral blood flow (CBF) is reduced in SVD, particularly within white matter.Here we quantified test-retest reliability in CBF measurements using pseudo-continuous arterial spin labelling (pCASL) in older adults with clinical and radiological evidence of SVD (N=54, mean (SD): 66.9 (8.7) years, 15 females/39 males). We generated whole-brain CBF maps on two visits at least 7 days apart (mean (SD): 20 (19), range 7-117 days).Test-retest reliability for CBF was high in all tissue types, with intra-class correlation coefficient [95%CI]: 0.758 [0.616, 0.852] for whole brain, 0.842 [0.743, 0.905] for total grey matter, 0.771 [0.636, 0.861] for deep grey matter (caudate-putamen and thalamus), 0.872 [0.790, 0.923] for normal-appearing white matter (NAWM) and 0.780 [0.650, 0.866] for WMH (all p<0.001). ANCOVA models indicated significant decline in CBF in total grey matter, deep grey matter and NAWM with increasing age and diastolic blood pressure (all p<0.001). CBF was lower in males relative to females (p=0.013 for total grey matter, p=0.004 for NAWM).We conclude that pCASL has high test-retest reliability as a quantitative measure of CBF in older adults with SVD. These findings support the use of pCASL in routine clinical imaging and as a clinical trial endpoint.All data come from the PASTIS trial, prospectively registered at: https://eudract.ema.europa.eu (2015-001235-20, registered 13/05/2015), http://www.clinicaltrials.gov (NCT02450253, registered 21/05/2015).
Collapse
Affiliation(s)
- Lauren R Binnie
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Mathilde M H Pauls
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Philip Benjamin
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
- Department of Neuroradiology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Mohani-Preet K Dhillon
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Shai Betteridge
- Department of Neuropsychology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Brian Clarke
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Rita Ghatala
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Fearghal A H Hainsworth
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Franklyn A Howe
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Usman Khan
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Christina Kruuse
- Department of Neurology and Neurovascular Research Unit, Herlev Gentofte Hospital, Herlev, Denmark
| | - Jeremy B Madigan
- Department of Neuroradiology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Barry Moynihan
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bhavini Patel
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Anthony C Pereira
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Egill Rostrup
- Mental Health Centre, University of Copenhagen, Glostrup, Denmark
| | - Anan B Y Shtaya
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Catherine A Spilling
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Sarah Trippier
- South London Stroke Research Network, St George's Hospital, London, UK
| | - Rebecca Williams
- South London Stroke Research Network, St George's Hospital, London, UK
| | - Jeremy D Isaacs
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK
| | - Thomas R Barrick
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Atticus H Hainsworth
- Molecular & Clinical Sciences Research Institute, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK.
- Department of Neurology, St George's University Hospitals NHS Foundation Trust London, London, UK.
| |
Collapse
|
32
|
He X, Dou W, Shi H. The Diagnostic Value of the Combined 3D Pseudo-Continuous Arterial Spin Labeling and Diffusion Kurtosis Imaging in Patients With Binswanger’s Disease. Front Neurosci 2022; 16:853422. [PMID: 35844226 PMCID: PMC9280636 DOI: 10.3389/fnins.2022.853422] [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: 01/12/2022] [Accepted: 06/09/2022] [Indexed: 12/04/2022] Open
Abstract
Background and Purpose The clinical diagnosis of Binswanger’s disease (BD), a chronic progressive form of subcortical vascular dementia, remains challenging. 3D pseudo-continuous arterial-spin-labeling (pcASL) and diffusion kurtosis imaging (DKI) can quantitatively reveal the microcirculation changes and heterogeneity of white matter (WM), respectively. We thus aimed to determine the diagnostic value of the combined 3D-pcASL and DKI in BD. Materials and Methods A total of 35 patients with BD and 33 healthy controls underwent 3D-ASL and DKI experiments. The perfusion parameter of cerebral blood flow (CBF), diffusion parameters of fractional anisotropy (FA), mean/axial/radial diffusivity (MD/Da/Dr), and kurtosis parameters of anisotropy fraction of kurtosis (FAk) and mean/axial/radial kurtosis MK/Ka/Kr were obtained to quantitatively measure the parametric distributions of functional brain subregions. One-way analysis of variance and post hoc t-test were applied to explore the different distributions of DKI/ASL-derived parameters among brain subregions of BD. In addition, all region-specific DKI/ASL parameters were separately analyzed in Pearson correlation analysis to investigate the relationship with Mini-Mental State Examination (MMSE), a typical clinical scale for cognitive function assessment in patients with BD. Results FA/FAk/MK/Ka/Kr was significantly declined in all WM hyperintensities (WMHs) of BD compared with healthy controls, while the corresponding MD/Da/Dr was significantly increased (all p < 0.005). In addition, significant changes, similar to the WMHs of patients with BD, were also observed in almost all DKI parameters in WM normal areas and genu/splenium of the corpus callosum (GCC/SCC) in BD (p < 0.005). Finally, CBF was significantly reduced in all of the above regions we measured in patients with BD (p < 0.005). For patients with BD, MMSE showed a negative correlation with MD/Da in thalamus (r = −0.42/−0.58; p < 0.05), and a positive correlation with CBF in PWM/TWM (r = 0.49/0.39; p < 0.05). Using receiver operating characteristic (ROC) analysis, FA/FAk/Kr in GCC, CBF/FA/Dr/FAk in SCC, MD/Da/Ka in thalamus, and the combined FA/MD/Dr/CBF in TWM showed high accuracy [area under the curves (AUCs) 0.957/0.946/0.942/0.986] in distinguishing patients with BD from healthy controls. Conclusion We found that combined DKI and 3D-ASL are helpful in diagnosing patients with BD, especially with FA, MD, Dr, and CBF in the temporal WM region. Additionally, the kurtosis parameters of DKI can sensitively monitor the potentially damaged WM areas in patients with BD patients, adding complementary clinical value.
Collapse
Affiliation(s)
- Xiaoyi He
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Radiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | | | - Hao Shi
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- *Correspondence: Hao Shi,
| |
Collapse
|
33
|
Zhou C, Sun P, Xu Y, Chen Y, Huang Y, Hamblin MH, Foley L, Hitchens TK, Li S, Yin K. Genetic Deficiency of MicroRNA-15a/16-1 Confers Resistance to Neuropathological Damage and Cognitive Dysfunction in Experimental Vascular Cognitive Impairment and Dementia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104986. [PMID: 35403823 PMCID: PMC9189640 DOI: 10.1002/advs.202104986] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/22/2022] [Indexed: 05/24/2023]
Abstract
Chronic cerebral hypoperfusion-derived brain damage contributes to the progression of vascular cognitive impairment and dementia (VCID). Cumulative evidence has shown that microRNAs (miRs) are emerging as novel therapeutic targets for CNS disorders. In this study, it is sought to determine the regulatory role of miR-15a/16-1 in VCID. It is found that miR-15a/16-1 knockout (KO) mice exhibit less cognitive and sensorimotor deficits following VCID. Genetic deficiency of miR-15a/16-1 in VCID mice also mitigate myelin degeneration, axonal injury, and neuronal loss. Mechanistically, miR-15a/16-1 binds to the 3'-UTR of AKT3 and IL-10RA. Genetic deletion of miR-15a/16-1 increases AKT3 and IL-10RA expression in VCID brains, and intranasal delivery of AKT3 and IL-10RA siRNA-loaded nanoparticles partially reduce brain protection and cognitive recovery in miR-15a/16-1 KO mice after VCID. In conclusion, the miR-15a/16-1-IL/10RA/AKT3 axis plays a critical role in regulating vascular brain damage and cognitive decline after VCID. Targeting miR-15a/16-1 is a novel therapeutic approach for the treatment of VCID.
Collapse
Affiliation(s)
- Chao Zhou
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Ping Sun
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Yuang Chen
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Yixian Huang
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Milton H. Hamblin
- Tulane University Health Sciences CenterTulane UniversityNew OrleansLA70112USA
| | - Lesley Foley
- Animal Imaging CenterDepartment of NeurobiologyUniversity of Pittsburgh School of MedicinePittsburghPA15203USA
| | - T. Kevin Hitchens
- Animal Imaging CenterDepartment of NeurobiologyUniversity of Pittsburgh School of MedicinePittsburghPA15203USA
| | - Song Li
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Ke‐Jie Yin
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPA15240USA
| |
Collapse
|
34
|
Abubakar MB, Sanusi KO, Ugusman A, Mohamed W, Kamal H, Ibrahim NH, Khoo CS, Kumar J. Alzheimer’s Disease: An Update and Insights Into Pathophysiology. Front Aging Neurosci 2022; 14:742408. [PMID: 35431894 PMCID: PMC9006951 DOI: 10.3389/fnagi.2022.742408] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/25/2022] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible brain disorder associated with slow, progressive loss of brain functions mostly in older people. The disease processes start years before the symptoms are manifested at which point most therapies may not be as effective. In the hippocampus, the key proteins involved in the JAK2/STAT3 signaling pathway, such as p-JAK2-Tyr1007 and p-STAT3-Tyr705 were found to be elevated in various models of AD. In addition to neurons, glial cells such as astrocytes also play a crucial role in the progression of AD. Without having a significant effect on tau and amyloid pathologies, the JAK2/STAT3 pathway in reactive astrocytes exhibits a behavioral impact in the experimental models of AD. Cholinergic atrophy in AD has been traced to a trophic failure in the NGF metabolic pathway, which is essential for the survival and maintenance of basal forebrain cholinergic neurons (BFCN). In AD, there is an alteration in the conversion of the proNGF to mature NGF (mNGF), in addition to an increase in degradation of the biologically active mNGF. Thus, the application of exogenous mNGF in experimental studies was shown to improve the recovery of atrophic BFCN. Furthermore, it is now coming to light that the FGF7/FGFR2/PI3K/Akt signaling pathway mediated by microRNA-107 is also involved in AD pathogenesis. Vascular dysfunction has long been associated with cognitive decline and increased risk of AD. Vascular risk factors are associated with higher tau and cerebral beta-amyloid (Aβ) burden, while synergistically acting with Aβ to induce cognitive decline. The apolipoprotein E4 polymorphism is not just one of the vascular risk factors, but also the most prevalent genetic risk factor of AD. More recently, the research focus on AD shifted toward metabolisms of various neurotransmitters, major and minor nutrients, thus giving rise to metabolomics, the most important “omics” tool for the diagnosis and prognosis of neurodegenerative diseases based on an individual’s metabolome. This review will therefore proffer a better understanding of novel signaling pathways associated with neural and glial mechanisms involved in AD, elaborate potential links between vascular dysfunction and AD, and recent developments in “omics”-based biomarkers in AD.
Collapse
Affiliation(s)
- Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Kamaldeen Olalekan Sanusi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wael Mohamed
- Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Malaysia
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Haziq Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Nurul Husna Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ching Soong Khoo
- Neurology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
- *Correspondence: Jaya Kumar,
| |
Collapse
|
35
|
Secondary Analysis of Walking Activities During the Acute Stroke Hospital Stay and Cerebrovascular Health. Cardiopulm Phys Ther J 2022; 33:130-137. [DOI: 10.1097/cpt.0000000000000196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Yang M, Sun D, Wang Y, Yan M, Zheng J, Ren J. Cognitive Impairment in Heart Failure: Landscape, Challenges, and Future Directions. Front Cardiovasc Med 2022; 8:831734. [PMID: 35198608 PMCID: PMC8858826 DOI: 10.3389/fcvm.2021.831734] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major global healthcare problem accounting for substantial deterioration of prognosis. As a complex clinical syndrome, HF often coexists with multi-comorbidities of which cognitive impairment (CI) is particularly important. CI is increasing in prevalence among patients with HF and is present in around 40%, even up to 60%, of elderly patients with HF. As a potent and independent prognostic factor, CI significantly increases the hospitalization and mortality and decreases quality of life in patients with HF. There has been a growing awareness of the complex bidirectional interaction between HF and CI as it shares a number of common pathophysiological pathways including reduced cerebral blood flow, inflammation, and neurohumoral activations. Research that focus on the precise mechanism for CI in HF is still ever insufficient. As the tremendous adverse consequences of CI in HF, effective early diagnosis of CI in HF and interventions for these patients may halt disease progression and improve prognosis. The current clinical guidelines in HF have begun to emphasize the importance of CI. However, nearly half of CI in HF is underdiagnosed, and few recommendations are available to guide clinicians about how to approach CI in patients with HF. This review aims to synthesize knowledge about the link between HF and cognitive dysfunction, issues pertaining to screening, diagnosis and management of CI in patients with HF, and emerging therapies for prevention. Based on data from current studies, critical gaps in knowledge of CI in HF are identified, and future research directions to guide the field forward are proposed.
Collapse
Affiliation(s)
- Mengxi Yang
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Di Sun
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Mengwen Yan
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingyi Ren
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
- Vascular Health Research Center of Peking University Health Science Center, Beijing, China
- *Correspondence: Jingyi Ren
| |
Collapse
|
37
|
Tu MC, Chung HW, Hsu YH, Yang JJ, Wu WC. Stage-Dependent Cerebral Blood Flow and Leukoaraiosis Couplings in Subcortical Ischemic Vascular Disease and Alzheimer's Disease. J Alzheimers Dis 2022; 86:729-739. [PMID: 35124651 PMCID: PMC9028753 DOI: 10.3233/jad-215405] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Alzheimer’s disease (AD) and subcortical ischemic vascular disease (SIVD) have both been associated with white matter hyperintensities (WMHs) and altered cerebral blood flow (CBF) although the etiology of AD is still unclear. Objective: To test the hypothesis that CBF and WMHs have differential effects on cognition and that the relationship between CBF and WMHs changes with the subtypes and stages of dementia. Methods: Forty-two patients with SIVD, 50 patients with clinically-diagnosed AD, and 30 cognitively-normal subjects were included. Based on the Clinical Dementia Rating (CDR), the patients were dichotomized into early-stage (CDR = 0.5) and late-stage (CDR = 1 or 2) groups. CBF and WMH metrics were derived from magnetic resonance imaging and correlated with cognition. Results: Hierarchical linear regression revealed that CBF metrics had distinct contribution to global cognition, memory, and attention, whereas WMH metrics had distinct contribution to executive function (all p < 0.05). In SIVD, the WMHs in frontotemporal areas correlated with the CBF in bilateral thalami at the early stage; the correlation then became between the WMHs in basal ganglia and the CBF in frontotemporal areas at the late stage. A similar corticosubcortical coupling was observed in AD but involved fewer areas. Conclusion: A stage-dependent coupling between CBF and WMHs was identified in AD and SIVD, where the extent of cortical WMHs correlated with subcortical CBF for CDR = 0.5, whereas the extent of subcortical WMHs correlated with cortical CBF for CDR = 1–2.
Collapse
Affiliation(s)
- Min-Chien Tu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.,Department of Neurology, Taichung Tzu Chi Hospital, Taichung, Taiwan.,Department of Neurology, Tzu Chi University, Hualien, Taiwan
| | - Hsiao-Wen Chung
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Yen-Hsuan Hsu
- Department of Psychology, National Chung Cheng University, Chiayi, Taiwan.,Center for Innovative Research on Aging Society, National Chung Cheng University, Chiayi, Taiwan
| | - Jir-Jei Yang
- Department of Radiology, Taichung Tzu Chi Hospital, Taichung, Taiwan
| | - Wen-Chau Wu
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.,Institute of Medical Device and Imaging, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
38
|
Xiang K, Liu Y, Sun L. Motoric Cognitive Risk Syndrome: Symptoms, Pathology, Diagnosis, and Recovery. Front Aging Neurosci 2022; 13:728799. [PMID: 35185512 PMCID: PMC8847709 DOI: 10.3389/fnagi.2021.728799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
The motoric cognitive risk (MCR) syndrome is a pre-dementia condition, marked by the enhanced risk for Alzheimer's disease (AD) and vascular dementia, together with falls, disability, and abnormal movements. The research studies revealed the distinct neurological and non-neurological clinical gait irregularities during dementia and accelerated functional decline, such as postural and balance impairments, memory loss, cognitive failure, and metabolic dysfunctions. The disabling characteristics of MCR comprise altered afferent sensory and efferent motor responses, together with disrupted visual, vestibular, and proprioceptive components. The pathological basis of MCR relates with the frontal lacunar infarcts, white matter hyperintensity (WMH), gray matter atrophy in the pre-motor and pre-frontal cortex, abnormal cholinergic functioning, inflammatory responses, and genetic factors. Further, cerebrovascular lesions and cardiovascular disorders exacerbate the disease pathology. The diagnosis of MCR is carried out through neuropsychological tests, biomarker assays, imaging studies, questionnaire-based evaluation, and motor function tests, including walking speed, dual-task gait tests, and ambulation ability. Recovery from MCR may include cognitive, physical, and social activities, exercise, diet, nutritional supplements, symptomatic drug treatment, and lifestyle habits that restrict the disease progression. Psychotherapeutic counseling, anti-depressants, and vitamins may support motor and cognitive improvement, primarily through the restorative pathways. However, an in-depth understanding of the association of immobility, dementia, and cognitive stress with MCR requires additional clinical and pre-clinical studies. They may have a significant contribution in reducing MCR syndrome and the risk for dementia. Overall, the current review informs the vital connection between gait performance and cognition in MCR and highlights the usefulness of future research in the discernment and treatment of dementiating illness.
Collapse
|
39
|
Li M, Kitamura A, Beverley J, Koudelka J, Duncombe J, Lennen R, Jansen MA, Marshall I, Platt B, Wiegand UK, Carare RO, Kalaria RN, Iliff JJ, Horsburgh K. Impaired Glymphatic Function and Pulsation Alterations in a Mouse Model of Vascular Cognitive Impairment. Front Aging Neurosci 2022; 13:788519. [PMID: 35095472 PMCID: PMC8793139 DOI: 10.3389/fnagi.2021.788519] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Large vessel disease and carotid stenosis are key mechanisms contributing to vascular cognitive impairment (VCI) and dementia. Our previous work, and that of others, using rodent models, demonstrated that bilateral common carotid stenosis (BCAS) leads to cognitive impairment via gradual deterioration of the neuro-glial-vascular unit and accumulation of amyloid-β (Aβ) protein. Since brain-wide drainage pathways (glymphatic) for waste clearance, including Aβ removal, have been implicated in the pathophysiology of VCI via glial mechanisms, we hypothesized that glymphatic function would be impaired in a BCAS model and exacerbated in the presence of Aβ. Male wild-type and Tg-SwDI (model of microvascular amyloid) mice were subjected to BCAS or sham surgery which led to a reduction in cerebral perfusion and impaired spatial learning acquisition and cognitive flexibility. After 3 months survival, glymphatic function was evaluated by cerebrospinal fluid (CSF) fluorescent tracer influx. We demonstrated that BCAS caused a marked regional reduction of CSF tracer influx in the dorsolateral cortex and CA1-DG molecular layer. In parallel to these changes increased reactive astrogliosis was observed post-BCAS. To further investigate the mechanisms that may lead to these changes, we measured the pulsation of cortical vessels. BCAS impaired vascular pulsation in pial arteries in WT and Tg-SwDI mice. Our findings show that BCAS influences VCI and that this is paralleled by impaired glymphatic drainage and reduced vascular pulsation. We propose that these additional targets need to be considered when treating VCI.
Collapse
Affiliation(s)
- Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Akihiro Kitamura
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Department of Neurology, Shiga University of Medical Science, Otsu, Japan
| | - Joshua Beverley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Lennen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Maurits A Jansen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Ian Marshall
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bettina Platt
- School of Medicine, Medical Sciences and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Ulrich K Wiegand
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roxana O Carare
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, United States
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
40
|
Poh L, Sim WL, Jo DG, Dinh QN, Drummond GR, Sobey CG, Chen CLH, Lai MKP, Fann DY, Arumugam TV. The role of inflammasomes in vascular cognitive impairment. Mol Neurodegener 2022; 17:4. [PMID: 35000611 PMCID: PMC8744307 DOI: 10.1186/s13024-021-00506-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
There is an increasing prevalence of Vascular Cognitive Impairment (VCI) worldwide, and several studies have suggested that Chronic Cerebral Hypoperfusion (CCH) plays a critical role in disease onset and progression. However, there is a limited understanding of the underlying pathophysiology of VCI, especially in relation to CCH. Neuroinflammation is a significant contributor in the progression of VCI as increased systemic levels of the proinflammatory cytokine interleukin-1β (IL-1β) has been extensively reported in VCI patients. Recently it has been established that CCH can activate the inflammasome signaling pathways, involving NLRP3 and AIM2 inflammasomes that critically regulate IL-1β production. Given that neuroinflammation is an early event in VCI, it is important that we understand its molecular and cellular mechanisms to enable development of disease-modifying treatments to reduce the structural brain damage and cognitive deficits that are observed clinically in the elderly. Hence, this review aims to provide a comprehensive insight into the molecular and cellular mechanisms involved in the pathogenesis of CCH-induced inflammasome signaling in VCI.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K. P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| |
Collapse
|
41
|
White matter damage as a consequence of vascular dysfunction in a spontaneous mouse model of chronic mild chronic hypoperfusion with eNOS deficiency. Mol Psychiatry 2022; 27:4754-4769. [PMID: 35948662 PMCID: PMC9734049 DOI: 10.1038/s41380-022-01701-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Vascular cognitive impairment and dementia (VCID) is the second most common form of dementia after Alzheimer's disease (AD). Currently, the mechanistic insights into the evolution and progression of VCID remain elusive. White matter change represents an invariant feature. Compelling clinical neuroimaging and pathological evidence suggest a link between white matter changes and neurodegeneration. Our prior study detected hypoperfused lesions in mice with partial deficiency of endothelial nitric oxide (eNOS) at very young age, precisely matching to those hypoperfused areas identified in preclinical AD patients. White matter tracts are particularly susceptible to the vascular damage induced by chronic hypoperfusion. Using immunohistochemistry, we detected severe demyelination in the middle-aged eNOS-deficient mice. The demyelinated areas were confined to cortical and subcortical areas including the corpus callosum and hippocampus. The intensity of demyelination correlated with behavioral deficits of gait and associative recognition memory performances. By Evans blue angiography, we detected blood-brain barrier (BBB) leakage as another early pathological change affecting frontal and parietal cortex in eNOS-deficient mice. Sodium nitrate fortified drinking water provided to young and middle-aged eNOS-deficient mice completely prevented non-perfusion, BBB leakage, and white matter pathology, indicating that impaired endothelium-derived NO signaling may have caused these pathological events. Furthermore, genome-wide transcriptomic analysis revealed altered gene clusters most related to mitochondrial respiratory pathways selectively in the white matter of young eNOS-deficient mice. Using eNOS-deficient mice, we identified BBB breakdown and hypoperfusion as the two earliest pathological events, resulting from insufficient vascular NO signaling. We speculate that the compromised BBB and mild chronic hypoperfusion trigger vascular damage, along with oxidative stress and astrogliosis, accounting for the white matter pathological changes in the eNOS-deficient mouse model. We conclude that eNOS-deficient mice represent an ideal spontaneous evolving model for studying the earliest events leading to white matter changes, which will be instrumental to future therapeutic testing of drug candidates and for targeting novel/specific vascular mechanisms contributing to VCID and AD.
Collapse
|
42
|
Capillary function progressively deteriorates in prodromal Alzheimer's disease: A longitudinal MRI perfusion study. AGING BRAIN 2022; 2:100035. [PMID: 36908896 PMCID: PMC9997144 DOI: 10.1016/j.nbas.2022.100035] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/20/2022] Open
Abstract
Cardiovascular risk factors are associated with the development of Alzheimer's disease (AD), and increasing evidence suggests that cerebral microvascular dysfunction plays a vital role in the disease progression. Using magnetic resonance imaging, we investigated the two-year changes of the cerebral microvascular blood flow in 11 mild cognitively impaired (MCI) patients with prodromal AD compared to 12 MCI patients without evidence of AD and 10 cognitively intact age-matched controls. The pAD-MCI patients displayed widespread deterioration in microvascular cerebral perfusion associated with capillary dysfunction. No such changes were observed in the other two groups, suggesting that the dysfunction in capillary perfusion is linked to the AD pathophysiology. The observed capillary dysfunction may limit local oxygenation in AD leading to downstream β-amyloid aggregation, tau hyperphosphorylation, neuroinflammation and neuronal dysfunction. The findings are in agreement with the capillary dysfunction hypothesis of AD, suggesting that increasing heterogeneity of capillary blood flow is a primary pathological event in AD.
Collapse
|
43
|
Mossanen Parsi M, Duval C, Ariëns RAS. Vascular Dementia and Crosstalk Between the Complement and Coagulation Systems. Front Cardiovasc Med 2021; 8:803169. [PMID: 35004913 PMCID: PMC8733168 DOI: 10.3389/fcvm.2021.803169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 01/12/2023] Open
Abstract
Vascular Dementia (VaD) is a neurocognitive disorder caused by reduced blood flow to the brain tissue, resulting in infarction, and is the second most common type of dementia. The complement and coagulation systems are evolutionary host defence mechanisms activated by acute tissue injury to induce inflammation, clot formation and lysis; recent studies have revealed that these systems are closely interlinked. Overactivation of these systems has been recognised to play a key role in the pathogenesis of neurological disorders such as Alzheimer's disease and multiple sclerosis, however their role in VaD has not yet been extensively reviewed. This review aims to bridge the gap in knowledge by collating current understanding of VaD to enable identification of complement and coagulation components involved in the pathogenesis of this disorder that may have their effects amplified or supressed by crosstalk. Exploration of these mechanisms may unveil novel therapeutic targets or biomarkers that would improve current treatment strategies for VaD.
Collapse
Affiliation(s)
| | | | - Robert A. S. Ariëns
- Discovery and Translational Science Department, School of Medicine, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
44
|
Diagnostic Efficacy of Voxel-Mirrored Homotopic Connectivity in Vascular Dementia as Compared to Alzheimer's Related Neurodegenerative Diseases-A Resting State fMRI Study. Life (Basel) 2021; 11:life11101108. [PMID: 34685479 PMCID: PMC8538280 DOI: 10.3390/life11101108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/10/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022] Open
Abstract
Previous studies have demonstrated that functional connectivity (FC) of different brain regions in resting state function MRI were abnormal in patients suffering from mild cognitive impairment (MCI) and Alzheimer’s disease (AD) when comparing to healthy controls (HC) using seed based, independent component analysis (ICA) or small world network techniques. A new technique called voxel-mirrored homotopic connectivity (VMHC) was used in the current study to evaluate the value of interhemispheric functional connectivity (IFC) as a diagnostic tool to differentiate vascular dementia (VD) from other Alzheimer’s related neurodegenerative diseases. Eighty-three participants were recruited from the university hospital memory clinic. A multidisciplinary panel formed by a neuroradiologist and two geriatricians classified the participants into VD (13), AD (16), MCI (29), and HC (25) based on clinical history, Montreal Cognitive Assessment Hong Kong version (HK-MoCA) neuropsychological score, structural MRI, MR perfusion, and 18-F Flutametamol (amyloid) PET-CT findings of individual subjects. We adopted the calculation method used by Kelly et al. (2011) and Zuo et al. (2010) in obtaining VMHC maps. Specific patterns of VMHC maps were obtained for VD, AD, and MCI to HC comparison. VD showed significant reduction in VMHC in frontal orbital gyrus and gyrus rectus. Increased VMHC was observed in default mode network (DMN), executive control network (ECN), and the remaining salient network (SN) regions. AD showed a reduction of IFC in all DMN, ECN, and SN regions; whereas MCI showed VMHC reduction in vSN, and increased VMHC in DMN and ECN. When combining VMHC values of relevant brain regions, the accuracy was improved to 87%, 92%, and 83% for VD, AD, and MCI from HC, respectively, in receiver operating characteristic (ROC) analysis. Through studying the VMHC maps and using VMHC values in relevant brain regions, VMHC can be considered as a reliable diagnostic tool for VD, AD, and MCI from HC.
Collapse
|
45
|
Hambali A, Kumar J, Hashim NFM, Maniam S, Mehat MZ, Cheema MS, Mustapha M, Adenan MI, Stanslas J, Hamid HA. Hypoxia-Induced Neuroinflammation in Alzheimer's Disease: Potential Neuroprotective Effects of Centella asiatica. Front Physiol 2021; 12:712317. [PMID: 34721056 PMCID: PMC8551388 DOI: 10.3389/fphys.2021.712317] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterised by the presence of extracellular beta-amyloid fibrillary plaques and intraneuronal neurofibrillary tau tangles in the brain. Recurring failures of drug candidates targeting these pathways have prompted research in AD multifactorial pathogenesis, including the role of neuroinflammation. Triggered by various factors, such as hypoxia, neuroinflammation is strongly linked to AD susceptibility and/or progression to dementia. Chronic hypoxia induces neuroinflammation by activating microglia, the resident immune cells in the brain, along with an increased in reactive oxygen species and pro-inflammatory cytokines, features that are common to many degenerative central nervous system (CNS) disorders. Hence, interests are emerging on therapeutic agents and plant derivatives for AD that target the hypoxia-neuroinflammation pathway. Centella asiatica is one of the natural products reported to show neuroprotective effects in various models of CNS diseases. Here, we review the complex hypoxia-induced neuroinflammation in the pathogenesis of AD and the potential application of Centella asiatica as a therapeutic agent in AD or dementia.
Collapse
Affiliation(s)
- Aqilah Hambali
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sandra Maniam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Manraj Singh Cheema
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Johnson Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
46
|
Liu S, Hou B, You H, Zhang Y, Zhu Y, Ma C, Zuo Z, Feng F. The Association Between Perivascular Spaces and Cerebral Blood Flow, Brain Volume, and Cardiovascular Risk. Front Aging Neurosci 2021; 13:599724. [PMID: 34531732 PMCID: PMC8438293 DOI: 10.3389/fnagi.2021.599724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Basal ganglia perivascular spaces are associated with cognitive decline and cardiovascular risk factors. There is a lack of studies on the cardiovascular risk burden of basal ganglia perivascular spaces (BG-PVS) and their relationship with gray matter volume (GMV) and GM cerebral blood flow (CBF) in the aging brain. Here, we investigated these two issues in a large sample of cognitively intact older adults. Methods: A total of 734 volunteers were recruited. MRI was performed with 3.0 T using a pseudo-continuous arterial spin labeling (pCASL) sequence and a sagittal isotropic T1-weighted sequence for CBF and GMV analysis. The images obtained from 406 participants were analyzed to investigate the relationship between the severity of BG-PVS and GMV/CBF. False discovery rate-corrected P-values (PFDR) of <0.05 were considered significant. The images obtained from 254 participants were used to study the relationship between the severity of BG-PVS and cardiovascular risk burden. BG-PVS were rated using a 5-grade score. The severity of BG-PVS was classified as mild (grade <3) and severe (grade ≥3). Cardiovascular risk burden was assessed with the Framingham General Cardiovascular Risk Score (FGCRS). Results: Severe basal ganglia perivascular spaces were associated with significantly smaller GMV and CBF in multiple cortical regions (PFDR <0.05), and were associated with significantly larger volume in the bilateral caudate nucleus, pallidum, and putamen (PFDR <0.05). The participants with severe BG-PVS were more likely to have a higher cardiovascular risk burden than the participants with mild BG-PVS (60.71% vs. 42.93%; P =0.02). Conclusion: In cognitively intact older adults, severe BG-PVS are associated with smaller cortical GMV and CBF, larger subcortical GMV, and higher cardiovascular risk burden.
Collapse
Affiliation(s)
- Sirui Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwei Zhang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Wang Y, Lu P, Zhan Y, Wu X, Qiu Y, Wang Z, Xu Q, Zhou Y. The Contribution of White Matter Diffusion and Cortical Perfusion Pathology to Vascular Cognitive Impairment: A Multimode Imaging-Based Machine Learning Study. Front Aging Neurosci 2021; 13:687001. [PMID: 34426730 PMCID: PMC8379092 DOI: 10.3389/fnagi.2021.687001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/02/2021] [Indexed: 11/13/2022] Open
Abstract
Widespread impairments in white matter and cerebrovascular integrity have been consistently implicated in the pathophysiology of patients with small vessel disease (SVD). However, the neural circuit mechanisms that underlie the developing progress of clinical cognitive symptoms remain largely elusive. Here, we conducted cross-modal MRI scanning including diffusion tensor imaging and arterial spin labeling in a cohort of 113 patients with SVD, which included 74 patients with vascular mild cognitive impairment (vMCI) and 39 patients without vMCI symptoms, and hence developed multimode imaging-based machine learning models to identify markers that discriminated SVD subtypes. Diffusion and perfusion features, respectively, extracted from individual white matter and gray matter regions were used to train three sets of classifiers in a nested 10-fold fashion: diffusion-based, perfusion-based, and combined diffusion-perfusion-based classifiers. We found that the diffusion-perfusion combined classifier achieved the highest accuracy of 72.57% with leave-one-out cross-validation, with the diffusion features largely spanning the capsular lateral pathway of the cholinergic tracts, and the perfusion features mainly distributed in the frontal-subcortical-limbic areas. Furthermore, diffusion-based features within vMCI group were associated with performance on executive function tests. We demonstrated the superior accuracy of using diffusion-perfusion combined multimode imaging features for classifying vMCI subtype out of a cohort of patients with SVD. Disruption of white matter integrity might play a critical role in the progression of cognitive impairment in patients with SVD, while malregulation of coritcal perfusion needs further study.
Collapse
Affiliation(s)
- Yao Wang
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peiwen Lu
- Department of Neurology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yafeng Zhan
- Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Xiaowei Wu
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yage Qiu
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Wang
- Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Qun Xu
- Department of Neurology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhou
- Department of Radiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Bandyopadhyay S. Role of Neuron and Glia in Alzheimer's Disease and Associated Vascular Dysfunction. Front Aging Neurosci 2021; 13:653334. [PMID: 34211387 PMCID: PMC8239194 DOI: 10.3389/fnagi.2021.653334] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloidogenicity and vascular dysfunction are the key players in the pathogenesis of Alzheimer’s disease (AD), involving dysregulated cellular interactions. An intricate balance between neurons, astrocytes, microglia, oligodendrocytes and vascular cells sustains the normal neuronal circuits. Conversely, cerebrovascular diseases overlap neuropathologically with AD, and glial dyshomeostasis promotes AD-associated neurodegenerative cascade. While pathological hallmarks of AD primarily include amyloid-β (Aβ) plaques and neurofibrillary tangles, microvascular disorders, altered cerebral blood flow (CBF), and blood-brain barrier (BBB) permeability induce neuronal loss and synaptic atrophy. Accordingly, microglia-mediated inflammation and astrogliosis disrupt the homeostasis of the neuro-vascular unit and stimulate infiltration of circulating leukocytes into the brain. Large-scale genetic and epidemiological studies demonstrate a critical role of cellular crosstalk for altered immune response, metabolism, and vasculature in AD. The glia associated genetic risk factors include APOE, TREM2, CD33, PGRN, CR1, and NLRP3, which correlate with the deposition and altered phagocytosis of Aβ. Moreover, aging-dependent downregulation of astrocyte and microglial Aβ-degrading enzymes limits the neurotrophic and neurogenic role of glial cells and inhibits lysosomal degradation and clearance of Aβ. Microglial cells secrete IGF-1, and neurons show a reduced responsiveness to the neurotrophic IGF-1R/IRS-2/PI3K signaling pathway, generating amyloidogenic and vascular dyshomeostasis in AD. Glial signals connect to neural stem cells, and a shift in glial phenotype over the AD trajectory even affects adult neurogenesis and the neurovascular niche. Overall, the current review informs about the interaction of neuronal and glial cell types in AD pathogenesis and its critical association with cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
49
|
Wang X, Yang X, Han F, Gao L, Zhou Y. Propofol improves brain injury induced by chronic cerebral hypoperfusion in rats. Food Sci Nutr 2021; 9:2801-2809. [PMID: 34136148 PMCID: PMC8194753 DOI: 10.1002/fsn3.1915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
To study effect of propofol on cognitive dysfunction and brain injury in a rat model of chronic cerebral hypoperfusion. The bilateral carotid artery ligation (bilateral common carotid artery occlusion and BCCAO) to establish rat model of chronic cerebral hypoperfusion and randomly assigned to 4 groups (n = 10): sham-operation group treated with saline model group, propofol treatment model group, normal saline treatment, propofol treatment in the sham-operation group; continuous intraperitoneal injection of propofol and saline for 12 weeks. Morris water maze was used to evaluate the learning and memory ability of rats. Determination of central cholinergic and oxidative stress in brain tissue by spectrophotometry. Detection of inflammatory response in brain tissue by immunohistochemistry and ELISA method. Detection of neuronal loss in brain tissue by Nissl and TUNEL staining. Compared with the saline-treated model group, propofol in model group significantly increased the rat brain tissue SOD activity (p < .01) and GPX activity (p < .01), decreased the MDA levels (p < .01) and protein carbonyl compound levels (p < .01). The propofol treatment of model group rats hippocampal GFAP-immunoreactive satellite glial cells (p < .01) and immune Iba1-positive microglia cells (p < .01) area percent compared to saline-treated model group decreased significantly. The number of normal propofol treatment of model group rats hippocampus neuron than in physiological saline treatment model group rats was significantly increased (p < .01). Propofol can improve chronic cerebral hypoperfusion in rats induced by cognitive dysfunction and brain damage.
Collapse
Affiliation(s)
- Xiaodong Wang
- Department of AnesthesiologyPeking University Hospital of StomatologyBeijingChina
| | - Xudong Yang
- Department of AnesthesiologyPeking University Hospital of StomatologyBeijingChina
| | - Fang Han
- Department of AnesthesiologyPeking University Hospital of StomatologyBeijingChina
| | - Ling Gao
- Department of AnesthesiologyPeking University Hospital of StomatologyBeijingChina
| | - Yi Zhou
- Department of AnesthesiologyPeking University Hospital of StomatologyBeijingChina
| |
Collapse
|
50
|
Pahlavian SH, Wang X, Ma S, Zheng H, Casey M, D’Orazio LM, Shao X, Ringman JM, Chui H, Wang DJJ, Yan L. Cerebroarterial pulsatility and resistivity indices are associated with cognitive impairment and white matter hyperintensity in elderly subjects: A phase-contrast MRI study. J Cereb Blood Flow Metab 2021; 41:670-683. [PMID: 32501154 PMCID: PMC7922759 DOI: 10.1177/0271678x20927101] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 01/22/2023]
Abstract
Increased cerebroarterial pulsations are thought to be contributing factors in microvascular damage and cognitive impairment. In this study, we assessed the utility of two-dimensional (2D) phase-contrast MRI (PC-MRI) in quantifying cerebroarterial pulsations and evaluated the associations of pulsatile and non-pulsatile hemodynamic measures with cognitive performance and white matter hyperintensities (WMH). Neurocognitive assessments on 50 elderly subjects were performed using clinical dementia rating (CDR) and Montreal cognitive assessment (MoCA). An electrocardiogram-gated 2D PC-MRI sequence was used to calculate mean flow rate, pulsatility index (PI), and resistivity index (RI) of the internal carotid artery. For each subject, whole brain global cerebral blood flow (gCBF) and relative WMH volume were also quantified. Elevated RI was significantly associated with reduced cognitive performance quantified using MoCA (p = 0.04) and global CDR (p = 0.02). PI and RI were both significantly associated with relative WMH volume (p = 0.01, p < 0.01, respectively). However, non-pulsatile hemodynamic measures were not associated with cognitive impairment or relative WMH volume. This study showed that the cerebroarterial pulsatile measures obtained using PC-MRI have stronger association with the measures of cognitive impairment compared to global blood flow measurement and as such, might be useful as potential biomarkers of cerebrovascular dysfunction in preclinical populations.
Collapse
Affiliation(s)
- Soroush H Pahlavian
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Xinhui Wang
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Samantha Ma
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Hong Zheng
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
| | - Marlena Casey
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Lina M D’Orazio
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Xingfeng Shao
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Helena Chui
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Danny JJ Wang
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| | - Lirong Yan
- USC Stevens Neuroimaging and
Informatics Institute, Keck School of Medicine, University of Southern
California, Los Angeles, CA, USA
- Department of Neurology, University
of Southern California, Los Angeles, CA, USA
| |
Collapse
|