1
|
Wang J, Meng X, Yang J, Tang Y, Zeng F, Wang Y, Chen Z, Chen D, Zou R, Liu W. Improvements in Exercise for Alzheimer's Disease: Highlighting FGF21-Induced Cerebrovascular Protection. Neurochem Res 2025; 50:95. [PMID: 39903342 DOI: 10.1007/s11064-025-04350-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Currently, it has shown a trend of earlier onset, with most patients experiencing a progressive decline in cognitive function following the disease's onset, which places a heavy burden on society and family. Since no drug cure for AD exists, exploring new ways for its treatment and prevention has become critical. Early vascular damage is an initial trigger for neuronal injury in AD, underscoring the importance of vascular health in the early stages of the disease. Patients with early AD experience abnormal blood-brain barrier transport of amyloid-β (Aβ) peptides, with excess Aβ being deposited in the cerebral vasculature. The toxic effects of Aβ lead to abnormalities in cerebrovascular structure and function. Fibroblast growth factor21 (FGF21) is an endocrine factor that positively regulates energy homeostasis and glucose-lipid metabolism. Notably, it is one of the effective targets for metabolic disease prevention and treatment. Recent studies have found that FGF21 has anti-aging and vasoprotective effects, with receptors for FGF21 present in the brain. Exercise stimulates the liver to produce large amounts of FGF21, which enters the blood-brain barrier with the blood to exert neurovascular protection. Therefore, we review the biological properties of FGF21, its role in the cerebrovascular structure and function in AD, and the mechanism of exercise-regulated FGF21 action on AD-related cerebrovascular changes, aiming to provide a new theoretical basis for using exercise to ameliorate degenerative neurological diseases.
Collapse
Affiliation(s)
- Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Jialun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Yingzhe Tang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Dandan Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, No. 437, Lushan South Road, Changsha, Hunan Province, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology, Ministry of Education, Hunan Normal University, Changsha, Hunan Province, 410081, China.
- Physical Education College, Yuelu District, Hunan Normal University, Changsha, Hunan Province, 410081, China.
| |
Collapse
|
2
|
Gu H, Liu LL, Wu A, Yu Y, Emir U, Sawiak SJ, Territo PR, Farlow MR, Zheng W, Du Y. Lead Acetate Exposure and Cerebral Amyloid Accumulation: Mechanistic Evaluations in APP/PS1 Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:107004. [PMID: 39412896 PMCID: PMC11482597 DOI: 10.1289/ehp14384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The role of environmental factors in Alzheimer's disease (AD) pathogenesis remains elusive. Mounting evidence suggests that acute and past exposure to the environmental toxicant lead (Pb) is associated with longitudinal decline in cognitive function, brain atrophy, and greater brain β -amyloid (A β ) deposition. However, the nature of Pb-induced amyloid deposition and how it contributes to AD development remain unclear. OBJECTIVES This study investigates the role of Pb in the pathogenesis of cerebral amyloid angiopathy (CAA) and whether plasminogen activator inhibitor-1 (PAI-1) contributes to this process in the APP/PS1 mouse model. METHODS Female APP/PS1 mice at 8 wk of age were administered either 50 mg / kg Pb-acetate (PbAc) (i.e., 27 mg Pb / kg ) or an equivalent molar concentration of sodium acetate (NaAc) via oral gavage once daily for 8 wk. Amyloid deposition and vascular amyloid were determined by immunostaining. In addition, A β perivascular drainage, vascular binding assay, and microglial endocytosis were examined to determine underlying mechanisms. Furthermore, magnetic resonance imaging demyelination imaging was performed in vivo measure the level of demyelination. Finally, Y-maze and Morris water maze tests were assessed to evaluate the cognitive function of mice. RESULTS APP/PS1 mice (an AD mice model) exposed to PbAc demonstrated more vascular amyloid deposition less neocortical myelination, and lower cognitive function, as well as greater vascular binding to A β 40 , higher A β 40 / A β 42 ratios, strikingly lower A β 40 levels in the perivascular drainage, and microglial endocytosis. Importantly, exposure to a specific PAI-1 inhibitor, tiplaxtinin, which previously was reported to lower CAA pathology in mice, resulted in less CAA-related outcomes following PbAc exposure. DISCUSSION Our findings suggest that PbAc induced CAA/AD pathogenesis via the PAI-1 signaling in the APP/PS1 mouse model, and the inhibition of PAI-1 could be a potential therapeutic target for PbAc-mediated CAA/AD disorders. https://doi.org/10.1289/EHP14384.
Collapse
Affiliation(s)
- Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Luqing L. Liu
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Alanna Wu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yongqi Yu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Uzay Emir
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Stephen J. Sawiak
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Paul R. Territo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matin R. Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Suzuki K, Ataka T, Kimura N, Matsubara E. Cognitive Impairment and Early-onset Cerebral Amyloid Angiopathy in a Middle-aged Man with a History of Childhood Traumatic Brain Injury. Intern Med 2024; 63:2547-2550. [PMID: 38346743 PMCID: PMC11473271 DOI: 10.2169/internalmedicine.2681-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/12/2023] [Indexed: 09/18/2024] Open
Abstract
We herein report the a 42-year-old man with early-onset cerebral amyloid angiopathy (CAA) and a history of traumatic brain injury and neurosurgery in childhood. Computed tomography revealed cognitive impairment and recurrent lobar intracerebral hemorrhaging. Magnetic resonance imaging indicated cerebral microbleeds, and Pittsburgh compound B positron emission tomography detected brain amyloid deposition, mainly in the region of trauma and occipital lobes. Interestingly, the patient had no genetic predispositions or relevant family history. This case suggests that a single traumatic brain injury or neurosurgery in childhood can cause early-onset CAA.
Collapse
Affiliation(s)
- Kosuke Suzuki
- Department of Neurology, Oita University Faculty of Medicine, Japan
| | - Takuya Ataka
- Department of Neurology, Oita University Faculty of Medicine, Japan
| | - Noriyuki Kimura
- Department of Neurology, Oita University Faculty of Medicine, Japan
| | - Etsuro Matsubara
- Department of Neurology, Oita University Faculty of Medicine, Japan
| |
Collapse
|
4
|
Parmaksiz D, Kim Y. Navigating Central Oxytocin Transport: Known Realms and Uncharted Territories. Neuroscientist 2024:10738584241268754. [PMID: 39113465 DOI: 10.1177/10738584241268754] [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] [Indexed: 01/06/2025]
Abstract
Complex mechanisms govern the transport and action of oxytocin (Oxt), a neuropeptide and hormone that mediates diverse physiologic processes. While Oxt exerts site-specific and rapid effects in the brain via axonal and somatodendritic release, volume transmission via CSF and the neurovascular interface can act as an additional mechanism to distribute Oxt signals across distant brain regions on a slower timescale. This review focuses on modes of Oxt transport and action in the CNS, with particular emphasis on the roles of perivascular spaces, the blood-brain barrier (BBB), and circumventricular organs in coordinating the triadic interaction among circulating blood, CSF, and parenchyma. Perivascular spaces, critical conduits for CSF flow, play a pivotal role in Oxt diffusion and distribution within the CNS and reciprocally undergo Oxt-mediated structural and functional reconstruction. While the BBB modulates the movement of Oxt between systemic and cerebral circulation in a majority of brain regions, circumventricular organs without a functional BBB can allow for diffusion, monitoring, and feedback regulation of bloodborne peripheral signals such as Oxt. Recognition of these additional transport mechanisms provides enhanced insight into the systemic propagation and regulation of Oxt activity.
Collapse
Affiliation(s)
- Deniz Parmaksiz
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
5
|
Rivera-Rivera LA, Vikner T, Eisenmenger L, Johnson SC, Johnson KM. Four-dimensional flow MRI for quantitative assessment of cerebrospinal fluid dynamics: Status and opportunities. NMR IN BIOMEDICINE 2024; 37:e5082. [PMID: 38124351 PMCID: PMC11162953 DOI: 10.1002/nbm.5082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/03/2023] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Neurological disorders can manifest with altered neurofluid dynamics in different compartments of the central nervous system. These include alterations in cerebral blood flow, cerebrospinal fluid (CSF) flow, and tissue biomechanics. Noninvasive quantitative assessment of neurofluid flow and tissue motion is feasible with phase contrast magnetic resonance imaging (PC MRI). While two-dimensional (2D) PC MRI is routinely utilized in research and clinical settings to assess flow dynamics through a single imaging slice, comprehensive neurofluid dynamic assessment can be limited or impractical. Recently, four-dimensional (4D) flow MRI (or time-resolved three-dimensional PC with three-directional velocity encoding) has emerged as a powerful extension of 2D PC, allowing for large volumetric coverage of fluid velocities at high spatiotemporal resolution within clinically reasonable scan times. Yet, most 4D flow studies have focused on blood flow imaging. Characterizing CSF flow dynamics with 4D flow (i.e., 4D CSF flow) is of high interest to understand normal brain and spine physiology, but also to study neurological disorders such as dysfunctional brain metabolite waste clearance, where CSF dynamics appear to play an important role. However, 4D CSF flow imaging is challenged by the long T1 time of CSF and slower velocities compared with blood flow, which can result in longer scan times from low flip angles and extended motion-sensitive gradients, hindering clinical adoption. In this work, we review the state of 4D CSF flow MRI including challenges, novel solutions from current research and ongoing needs, examples of clinical and research applications, and discuss an outlook on the future of 4D CSF flow.
Collapse
Affiliation(s)
- Leonardo A Rivera-Rivera
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tomas Vikner
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiation Sciences, Radiation Physics and Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Laura Eisenmenger
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kevin M Johnson
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
6
|
Chen HC, Cao JX, Zhang YS, Ma YZ, Zhang L, Su XM, Gao LP, Jing YH. High salt diet exacerbates cognitive deficits and neurovascular abnormalities in APP/PS1 mice and induces AD-like changes in wild-type mice. J Nutr Biochem 2024; 125:109570. [PMID: 38218348 DOI: 10.1016/j.jnutbio.2024.109570] [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: 06/13/2023] [Revised: 12/25/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
High salt diet (HSD) is a risk factor of hypertension and cardiovascular disease. Although clinical data do not clearly indicate the relationship between HSD and the prevalence of Alzheimer's disease (AD), animal experiments have shown that HSD can cause hyperphosphorylation of tau protein and cognition impairment. However, whether HSD can accelerate the progression of AD by damaging the function of neurovascular unit (NVU) in the brain is unclear. Here, we fed APP/PS1 mice (an AD model) or wild-type mice with HSD and found that the chronic HSD feeding increased the activity of enzymes related to tau phosphorylation, which led to tau hyperphosphorylation in the brain. HSD also aggravated the deposition of Aβ42 in hippocampus and cortex in the APP/PS1 mice but not in the wild-type mice. Simultaneously, HSD caused the microglia proliferation, low expression of Aqp-4, and high expression of CD31 in the wild-type mice, which were accompanied with the loss of pericytes (PCs) and increase in blood brain barrier (BBB) permeability. As a result, wild-type mice fed with HSD performed poorly in Morris Water Maze and object recognition test. In the APP/PS1 mice, HSD feeding for 8 months worsen the cognition and accompanied the loss of PCs, the activation of glia, the increase in BBB permeability, and the acceleration of calcification in the brain. Our data suggested that HSD feeding induced the AD-like pathology in wild-type mice and aggravated the development of AD-like pathology in APP/PS1 mice, which implicated the tau hyperphosphorylation and NVU dysfunction.
Collapse
Affiliation(s)
- Hai Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Lu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, People's Republic of China; Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, Lanzhou, Gansu, People's Republic of China.
| |
Collapse
|
7
|
Royse SK, Snitz BE, Hengenius JB, Huppert TJ, Roush RE, Ehrenkranz RE, Wilson JD, Bertolet M, Reese AC, Cisneros G, Potopenko K, Becker JT, Cohen AD, Shaaban CE. Unhealthy white matter connectivity, cognition, and racialization in older adults. Alzheimers Dement 2024; 20:1483-1496. [PMID: 37828730 PMCID: PMC10947965 DOI: 10.1002/alz.13494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION White matter hyperintensities (WMH) may promote clinical Alzheimer's disease (AD) disparities between Black American (BA) and non-Hispanic White (nHW) populations. Using a novel measurement, unhealthy white matter connectivity (UWMC), we interrogated racialized group differences in associations between WMH in AD pathology-affected regions and cognition. METHODS UWMC is the proportion of white matter fibers that pass through WMH for every pair of brain regions. Individual regression models tested associations of UWMC in beta-amyloid (Aβ) or tau pathology-affected regions with cognition overall, stratified by racialized group, and with a racialized group interaction. RESULTS In 201 older adults ranging from cognitively unimpaired to AD, BA participants exhibited greater UWMC and worse cognition than nHW participants. UWMC was negatively associated with cognition in 17 and 5 Aβ- and tau-affected regions, respectively. Racialization did not modify these relationships. DISCUSSION Differential UWMC burden, not differential UWMC-and-cognition associations, may drive clinical AD disparities between racialized groups. HIGHLIGHTS Unhealthy white matter connectivity (UWMC) in Alzheimer's disease (AD) pathology-affected brain regions is associated with cognition. Relationships between UWMC and cognition are similar between Black American (BA) and non-Hispanic White (nHW) individuals. More UWMC may partially drive higher clinical AD burden in BA versus nHW populations. UWMC risk factors, particularly social and environmental, should be identified.
Collapse
Affiliation(s)
- Sarah K. Royse
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beth E. Snitz
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James B. Hengenius
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Theodore J. Huppert
- Department of Electrical EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rebecca E. Roush
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - James D. Wilson
- Department of Mathematics and StatisticsUniversity of San FranciscoSan FranciscoCaliforniaUSA
| | - Marnie Bertolet
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Geraldine Cisneros
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Katey Potopenko
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James T. Becker
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ann D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
8
|
Hong H, Hong L, Luo X, Zeng Q, Li K, Wang S, Jiaerken Y, Zhang R, Yu X, Zhang Y, Lei C, Liu Z, Chen Y, Huang P, Zhang M. The relationship between amyloid pathology, cerebral small vessel disease, glymphatic dysfunction, and cognition: a study based on Alzheimer's disease continuum participants. Alzheimers Res Ther 2024; 16:43. [PMID: 38378607 PMCID: PMC10877805 DOI: 10.1186/s13195-024-01407-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 02/04/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Glymphatic dysfunction is a crucial pathway for dementia. Alzheimer's disease (AD) pathologies co-existing with cerebral small vessel disease (CSVD) is the most common pathogenesis for dementia. We hypothesize that AD pathologies and CSVD could be associated with glymphatic dysfunction, contributing to cognitive impairment. METHOD Participants completed with amyloid PET, diffusion tensor imaging (DTI), and T2 fluid-attenuated inversion-recovery (FLAIR) sequences were included from the Alzheimer's Disease Neuroimaging Initiative (ADNI). White matter hyperintensities (WMH), the most common CSVD marker, was evaluated from T2FLAIR images and represented the burden of CSVD. Amyloid PET was used to assess Aβ aggregation in the brain. We used diffusion tensor image analysis along the perivascular space (DTI-ALPS) index, the burden of enlarged perivascular spaces (PVS), and choroid plexus volume to reflect glymphatic function. The relationships between WMH burden/Aβ aggregation and these glymphatic markers as well as the correlations between glymphatic markers and cognitive function were investigated. Furthermore, we conducted mediation analyses to explore the potential mediating effects of glymphatic markers in the relationship between WMH burden/Aβ aggregation and cognition. RESULTS One hundred and thirty-three participants along the AD continuum were included, consisting of 40 CN - , 48 CN + , 26 MCI + , and 19 AD + participants. Our findings revealed that there were negative associations between whole-brain Aβ aggregation (r = - 0.249, p = 0.022) and WMH burden (r = - 0.458, p < 0.001) with DTI-ALPS. Additionally, Aβ aggregation (r = 0.223, p = 0.041) and WMH burden (r = 0.294, p = 0.006) were both positively associated with choroid plexus volume. However, we did not observe significant correlations with PVS enlargement severity. DTI-ALPS was positively associated with memory (r = 0.470, FDR-p < 0.001), executive function (r = 0.358, FDR-p = 0.001), visual-spatial (r = 0.223, FDR-p < 0.040), and language (r = 0.419, FDR-p < 0.001). Conversely, choroid plexus volume showed negative correlations with memory (r = - 0.315, FDR-p = 0.007), executive function (r = - 0.321, FDR-p = 0.007), visual-spatial (r = - 0.233, FDR-p = 0.031), and language (r = - 0.261, FDR-p = 0.021). There were no significant correlations between PVS enlargement severity and cognitive performance. In the mediation analysis, we found that DTI-ALPS acted as a mediator in the relationship between WMH burden/Aβ accumulation and memory and language performances. CONCLUSION Our study provided evidence that both AD pathology (Aβ) and CSVD were associated with glymphatic dysfunction, which is further related to cognitive impairment. These results may provide a theoretical basis for new targets for treating AD.
Collapse
Affiliation(s)
- Hui Hong
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Luwei Hong
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Kaicheng Li
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Shuyue Wang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yeerfan Jiaerken
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Ruiting Zhang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xinfeng Yu
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yao Zhang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Cui Lei
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Zhirong Liu
- Department of Neurology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yanxing Chen
- Department of Neurology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China.
| | - Minming Zhang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Peng T, Xie Y, Liu F, Lian Y, Xie Y, Ma Y, Wang C, Xie N. The cerebral lymphatic drainage system and its implications in epilepsy. J Neurosci Res 2024; 102:e25267. [PMID: 38284855 DOI: 10.1002/jnr.25267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/18/2023] [Accepted: 10/08/2023] [Indexed: 01/30/2024]
Abstract
The central nervous system has long been thought to lack a clearance system similar to the peripheral lymphatic system. Therefore, the clearance of metabolic waste in the central nervous system has been a subject of great interest in neuroscience. Recently, the cerebral lymphatic drainage system, including the parenchymal clearance system and the meningeal lymphatic network, has attracted considerable attention. It has been extensively studied in various neurological disorders. Solute accumulation and neuroinflammation after epilepsy impair the blood-brain barrier, affecting the exchange and clearance between cerebrospinal fluid and interstitial fluid. Restoring their normal function may improve the prognosis of epilepsy. However, few studies have focused on providing a comprehensive overview of the brain clearance system and its significance in epilepsy. Therefore, this review addressed the structural composition, functions, and methods used to assess the cerebral lymphatic system, as well as the neglected association with epilepsy, and provided a theoretical basis for therapeutic approaches in epilepsy.
Collapse
Affiliation(s)
- Tingting Peng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, P.R. China
| | - Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, P.R. China
| | - Fengxia Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yunqing Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Cui Wang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
10
|
Loeffler DA. Approaches for Increasing Cerebral Efflux of Amyloid-β in Experimental Systems. J Alzheimers Dis 2024; 100:379-411. [PMID: 38875041 PMCID: PMC11307100 DOI: 10.3233/jad-240212] [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] [Accepted: 05/14/2024] [Indexed: 06/16/2024]
Abstract
Amyloid protein-β (Aβ) concentrations are increased in the brain in both early onset and late onset Alzheimer's disease (AD). In early onset AD, cerebral Aβ production is increased and its clearance is decreased, while increased Aβ burden in late onset AD is due to impaired clearance. Aβ has been the focus of AD therapeutics since development of the amyloid hypothesis, but efforts to slow AD progression by lowering brain Aβ failed until phase 3 trials with the monoclonal antibodies lecanemab and donanemab. In addition to promoting phagocytic clearance of Aβ, antibodies lower cerebral Aβ by efflux of Aβ-antibody complexes across the capillary endothelia, dissolving Aβ aggregates, and a "peripheral sink" mechanism. Although the blood-brain barrier is the main route by which soluble Aβ leaves the brain (facilitated by low-density lipoprotein receptor-related protein-1 and ATP-binding cassette sub-family B member 1), Aβ can also be removed via the blood-cerebrospinal fluid barrier, glymphatic drainage, and intramural periarterial drainage. This review discusses experimental approaches to increase cerebral Aβ efflux via these mechanisms, clinical applications of these approaches, and findings in clinical trials with these approaches in patients with AD or mild cognitive impairment. Based on negative findings in clinical trials with previous approaches targeting monomeric Aβ, increasing the cerebral efflux of soluble Aβ is unlikely to slow AD progression if used as monotherapy. But if used as an adjunct to treatment with lecanemab or donanemab, this approach might allow greater slowing of AD progression than treatment with either antibody alone.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, USA
| |
Collapse
|
11
|
Poliakova T, Wellington CL. Roles of peripheral lipoproteins and cholesteryl ester transfer protein in the vascular contributions to cognitive impairment and dementia. Mol Neurodegener 2023; 18:86. [PMID: 37974180 PMCID: PMC10652636 DOI: 10.1186/s13024-023-00671-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
This narrative review focuses on the role of cholesteryl ester transfer protein (CETP) and peripheral lipoproteins in the vascular contributions to cognitive impairment and dementia (VCID). Humans have a peripheral lipoprotein profile where low-density lipoproteins (LDL) represent the dominant lipoprotein fraction and high-density lipoproteins (HDL) represent a minor lipoprotein fraction. Elevated LDL-cholesterol (LDL-C) levels are well-established to cause cardiovascular disease and several LDL-C-lowering therapies are clinically available to manage this vascular risk factor. The efficacy of LDL-C-lowering therapies to reduce risk of all-cause dementia and AD is now important to address as recent studies demonstrate a role for LDL in Alzheimer's Disease (AD) as well as in all-cause dementia. The LDL:HDL ratio in humans is set mainly by CETP activity, which exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise LDL and lower HDL as CETP activity increases. Genetic and pharmacological studies support the hypothesis that CETP inhibition reduces cardiovascular risk by lowering LDL, which, by extension, may also lower VCID. Unlike humans, wild-type mice do not express catalytically active CETP and have HDL as their major lipoprotein fraction. As HDL has potent beneficial effects on endothelial cells, the naturally high HDL levels in mice protect them from vascular disorders, likely including VCID. Genetic restoration of CETP expression in mice to generate a more human-like lipid profile may increase the relevance of murine models for VCID studies. The therapeutic potential of existing and emerging LDL-lowering therapies for VCID will be discussed. Figure Legend. Cholesteryl Ester Transfer Protein in Alzheimer's Disease. CETP is mainly produced by the liver, and exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise circulating LDL and lower HDL as CETP activity increases. Low CETP activity is associated with better cardiovascular health, due to decreased LDL and increased HDL, which may also improve brain health. Although most peripheral lipoproteins cannot enter the brain parenchyma due to the BBB, it is increasingly appreciated that direct access to the vascular endothelium may enable peripheral lipoproteins to have indirect effects on brain health. Thus, lipoproteins may affect the cerebrovasculature from both sides of the BBB. Recent studies show an association between elevated plasma LDL, a well-known cardiovascular risk factor, and a higher risk of AD, and considerable evidence suggests that high HDL levels are associated with reduced CAA and lower neuroinflammation. Considering the potential detrimental role of LDL in AD and the importance of HDL's beneficial effects on endothelial cells, high CETP activity may lead to compromised BBB integrity, increased CAA deposits and greater neuroinflammation. Abbreviations: CETP - cholesteryl transfer ester protein; LDL - low-density lipoproteins; HDL - high-density lipoproteins; BBB - blood-brain barrier; CAA - cerebral amyloid angiopathy, SMC - smooth muscle cells, PVM - perivascular macrophages, RBC - red blood cells.
Collapse
Affiliation(s)
- Tetiana Poliakova
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- International Collaboration On Repair Discoveries, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Shi H, Koronyo Y, Fuchs DT, Sheyn J, Jallow O, Mandalia K, Graham SL, Gupta VK, Mirzaei M, Kramerov AA, Ljubimov AV, Hawes D, Miller CA, Black KL, Carare RO, Koronyo-Hamaoui M. Retinal arterial Aβ 40 deposition is linked with tight junction loss and cerebral amyloid angiopathy in MCI and AD patients. Alzheimers Dement 2023; 19:5185-5197. [PMID: 37166032 PMCID: PMC10638467 DOI: 10.1002/alz.13086] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION Vascular amyloid beta (Aβ) protein deposits were detected in retinas of mild cognitively impaired (MCI) and Alzheimer's disease (AD) patients. We tested the hypothesis that the retinal vascular tight junctions (TJs) were compromised and linked to disease status. METHODS TJ components and Aβ expression in capillaries and larger blood vessels were determined in post mortem retinas from 34 MCI or AD patients and 27 cognitively normal controls and correlated with neuropathology. RESULTS Severe decreases in retinal vascular zonula occludens-1 (ZO-1) and claudin-5 correlating with abundant arteriolar Aβ40 deposition were identified in MCI and AD patients. Retinal claudin-5 deficiency was closely associated with cerebral amyloid angiopathy, whereas ZO-1 defects correlated with cerebral pathology and cognitive deficits. DISCUSSION We uncovered deficiencies in blood-retinal barrier markers for potential retinal imaging targets of AD screening and monitoring. Intense retinal arteriolar Aβ40 deposition suggests a common pathogenic mechanism of failed Aβ clearance via intramural periarterial drainage.
Collapse
Affiliation(s)
- Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Krishna Mandalia
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stuart L. Graham
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Vivek K. Gupta
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Mehdi Mirzaei
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Andrei A. Kramerov
- Department of Biomedical Sciences and Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alexander V. Ljubimov
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences and Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Debra Hawes
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90048, USA
| | - Carol A. Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90048, USA
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Roxana O. Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton SO16 6YD, UK
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
13
|
Wang H, Zhang Z, Sittirattanayeunyong S, Hongpaisan J. Association of Apolipoprotein E4-related Microvascular Disease in the Alzheimer's Disease Hippocampal CA1 Stratum Radiatum. Neuroscience 2023; 526:204-222. [PMID: 37385335 PMCID: PMC10528415 DOI: 10.1016/j.neuroscience.2023.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
Current data suggest a hypothesis of vascular pathogenesis for the development and progression of Alzheimer's disease (AD). To investigate this, we studied the association of apolipoprotein E4 (APOE4) gene on microvessels in human autopsy-confirmed AD with and without APOE4, compared with age/sex-matched control (AC) hippocampal CA1 stratum radiatum. AD arterioles (without APOE4 gene) had mild oxidative stress and loss of vascular endothelial growth factor (VEGF) and endothelial cell density, reflecting aging progression. In AD + APOE4, an increase in strong oxidative DNA damage marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), VEGF, and endothelial cell density were associated with increased diameter of arterioles and perivascular space dilation. In cultured human brain microvascular cells (HBMECs), treatment of ApoE4 protein plus amyloid-β (Aβ) oligomers increased superoxide production and the apoptotic marker cleaved caspase 3, sustained hypoxia inducible factor-1α (HIF-1α) stability that was associated with an increase in MnSOD, VEGF, and cell density. This cell over-proliferation was inhibited with the antioxidants N-acetyl cysteine and MnTMPyP, the HIF-1α inhibitor echinomycin, the VEGFR-2 receptor blocker SU1498, the protein kinase C (PKC) ε knock-down (KD) and the extracellular signal-regulated kinase 1/2 (ERK) inhibitor FR180204. The PKCε KD and echinomycin decreased VEGF and/or ERK. In conclusion, AD capillaries and arterioles in hippocampal CA1 stratum radiatum of non-APOE4 carriers are related with aging, while those in APOE4 carriers with AD are related with pathogenesis of cerebrovascular disease.
Collapse
Affiliation(s)
- Huaixing Wang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Zongxiu Zhang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sorawit Sittirattanayeunyong
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
14
|
Miao M, Lyu M, Zhong C, Liu Y. Correlation Between MMP9 Promoter Methylation and Transient Ischemic Attack/Mild Ischemic Stroke with Early Cognitive Impairment. Clin Interv Aging 2023; 18:1221-1232. [PMID: 37547382 PMCID: PMC10404041 DOI: 10.2147/cia.s421830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/22/2023] [Indexed: 08/08/2023] Open
Abstract
Background/Objective Dyskinesia caused by transient ischemic attack (TIA) and mild ischemic stroke (MIS) is mild and short-lived; however, cognitive impairment (CI) can occur in the acute phase and be easily overlooked. DNA methylation is an epigenetic phenomenon that can affect gene expression through gene silencing. Blood levels of matrix metalloproteinase (MMP) 9 are elevated in ischemic stroke patients and is associated with the destruction of the blood-brain barrier and the occurrence of CI. No studies have investigated the relationship between MMP9 gene methylation and TIA/MIS with early cognitive impairment (ECI). As such, the purpose of the present study was to investigate the correlation between MMP9 gene methylation and TIA/MIS with ECI. Methods Data from 112 subjects were collected, including 84 with TIA/MIS (National Institutes of Health Stroke Scale <5 points) and 28 non-stroke control subjects. Patients were evaluated within 7 days of TIA/MIS onset according to four single-domain cognitive scales. Whole blood DNA methylation was detected using MethylTarget sequencing technology. Comparison of MMP9 gene methylation levels among subgroups was performed using statistical methods. Results The site S33-79 in the TIA/MIS group was hypomethylated compared with the control group, and sites S33-25 and S33-30 in TIA/MIS with ECI was hypomethylated compared with TIA/MIS without ECI. Compared with the small artery occlusion group, MMP9 gene, S33-25, 30, 39, 53, 58, 73, 79, 113 and 131 sites in the large artery atherosclerosis group were hypomethylated. Conclusion MMP9 gene hypomethylation sites were associated with TIA/MIS and TIA/MIS with ECI, and there was a strong correlation between MMP9 gene hypomethylation and atherosclerotic TIA/MIS. MMP9 gene methylation can reflect the severity of TIA/MIS. MMP9 gene hypomethylation sites may be used as potential biomarkers and therapeutic targets for TIA/MIS and TIA/MIS with ECI.
Collapse
Affiliation(s)
- Meng Miao
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, People’s Republic of China
| | - Mingyang Lyu
- Haihe Laboratory of Cell Ecosystem, Tianjin, People’s Republic of China
| | - Chi Zhong
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, People’s Republic of China
| | - Ying Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
15
|
Storti B, Gabriel MM, Sennfält S, Canavero I, Rifino N, Gatti L, Bersano A. Rare forms of cerebral amyloid angiopathy: pathogenesis, biological and clinical features of CAA-ri and iCAA. Front Neurosci 2023; 17:1219025. [PMID: 37492402 PMCID: PMC10363735 DOI: 10.3389/fnins.2023.1219025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 07/27/2023] Open
Abstract
Thanks to a more widespread knowledge of the disease, and improved diagnostic techniques, the clinical spectrum of cerebral amyloid angiopathy (CAA) is now broad. Sporadic CAA, hereditary CAA, CAA-related inflammation (CAA-ri) and iatrogenic CAA (iCAA) create a clinical and radiological continuum which is intriguing and only partially discovered. Despite being relatively rare, CAA-ri, an aggressive subtype of CAA with vascular inflammation, has gained growing attention also because of the therapeutic efficacy of anti-inflammatory and immunomodulating drugs. More recently, diagnostic criteria have been proposed for an unusual variant of CAA, probably related to an iatrogenic origin (iCAA), toward which there is mounting scientific interest. These atypical forms of CAA are still poorly known, and their recognition can be challenging and deserve to be pursued in specialized referral centres. The aim of this brief review is to focus current developments in the field of rare forms of CAA, its pathogenesis as well as clinical and biological features in order to increase awareness of these rare forms.
Collapse
Affiliation(s)
- Benedetta Storti
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Magdalena Gabriel
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Stefan Sennfält
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Isabella Canavero
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nicola Rifino
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Gatti
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
16
|
Xu J, Su Y, Fu J, Shen Y, Dong Q, Cheng X. Glymphatic pathway in sporadic cerebral small vessel diseases: From bench to bedside. Ageing Res Rev 2023; 86:101885. [PMID: 36801378 DOI: 10.1016/j.arr.2023.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Cerebral small vessel diseases (CSVD) consist of a group of diseases with high heterogeneity induced by pathologies of intracranial small blood vessels. Endothelium dysfunction, bloodbrain barrier leakage and the inflammatory response are traditionally considered to participate in the pathogenesis of CSVD. However, these features cannot fully explain the complex syndrome and related neuroimaging characteristics. In recent years, the glymphatic pathway has been discovered to play a pivotal role in clearing perivascular fluid and metabolic solutes, which has provided novel insights into neurological disorders. Researchers have also explored the potential role of perivascular clearance dysfunction in CSVD. In this review, we presented a brief overview of CSVD and the glymphatic pathway. In addition, we elucidated CSVD pathogenesis from the perspective of glymphatic failure, including basic animal models and clinical neuroimaging markers. Finally, we proposed forthcoming clinical applications targeting the glymphatic pathway, hoping to provide novel ideas on promising therapies and preventions of CSVD.
Collapse
Affiliation(s)
- Jiajie Xu
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Su
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayu Fu
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC and Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiang Dong
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Matsuzono K, Suzuki M, Anan Y, Ozawa T, Mashiko T, Koide R, Tanaka R, Fujimoto S. Spontaneous Echo Contrast in the Left Atrium and Aortic-Arch Atheroma, Detected by Transesophageal Echocardiography, Was Negatively Correlated with Cognitive Function. J Alzheimers Dis 2023; 91:673-681. [PMID: 36463447 DOI: 10.3233/jad-220763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND The relationship between transesophageal echocardiography findings and cognitive function. OBJECTIVE This study aimed to establish an association between transesophageal echocardiography findings and cognitive function in stroke survivors. METHODS A single-center study was conducted between April 1, 2017 and March 31, 2022. All subjects that were included had a past history of ischemic stroke and were admitted after >21 days from onset. The participants underwent cognitive function tests including a Mini-Mental State Examination, Revised Hasegawa Dementia Scale, Frontal Assessment Battery, and transesophageal echocardiography. RESULTS The results of 126 participants were analyzed. The cognitive function of participants with a spontaneous echo contrast (+) in the left atrium including appendage or of those with an aorta-arch plaque with a maximum thickness ≥4 mm significantly worse while neither the patent foramen ovale nor the branch extending plaque influenced cognitive function (The median cognitive scores of the spontaneous echo contrast (-) versus (+) were 26 versus 22, p < 0.01**, 26 versus 21, p < 0.001***, and 14 versus 11, p < 0.01**. Those of the aortic-arch plaque max thickness (<4 mm) versus (≥4 mm) were 26 versus 25, p < 0.05*, 27 versus 24, p < 0.05*, and 15 versus 13, p < 0.05*). CONCLUSION Our findings show that spontaneous echo contrast in the left atrium and aortic-arch atheroma detected by transesophageal echocardiography, were negatively associated with cognitive function.
Collapse
Affiliation(s)
- Kosuke Matsuzono
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masayuki Suzuki
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Yuhei Anan
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tadashi Ozawa
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takafumi Mashiko
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Reiji Koide
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ryota Tanaka
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shigeru Fujimoto
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
18
|
van der Thiel MM, Backes WH, Ramakers IHGB, Jansen JFA. Novel developments in non-contrast enhanced MRI of the perivascular clearance system: What are the possibilities for Alzheimer's disease research? Neurosci Biobehav Rev 2023; 144:104999. [PMID: 36529311 DOI: 10.1016/j.neubiorev.2022.104999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
The cerebral waste clearance system (i.e, glymphatic or intramural periarterial drainage) works through a network of perivascular spaces (PVS). Dysfunction of this system likely contributes to aggregation of Amyloid-β and subsequent toxic plaques in Alzheimer's disease (AD). A promising, non-invasive technique to study this system is MRI, though applications in dementia are still scarce. This review focusses on recent non-contrast enhanced (non-CE) MRI techniques which determine and visualise physiological aspects of the clearance system at multiple levels, i.e., cerebrospinal fluid flow, PVS-flow and interstitial fluid movement. Furthermore, various MRI studies focussing on aspects of the clearance system which are relevant to AD are discussed, such as studies on ageing, sleep alterations, and cognitive decline. Additionally, the complementary function of non-CE to CE methods is elaborated upon. We conclude that non-CE studies have great potential to determine which parts of the waste clearance system are affected by AD and in which stages of cognitive impairment dysfunction of this system occurs, which could allow future clinical trials to target these specific mechanisms.
Collapse
Affiliation(s)
- Merel M van der Thiel
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Inez H G B Ramakers
- Department of Psychiatry &Neuropsychology, Maastricht University, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; School for Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
19
|
Yan JH, Wang YJ, Sun YR, Pei YH, Ma HW, Mu YK, Qin LH. The lymphatic drainage systems in the brain: a novel target for ischemic stroke? Neural Regen Res 2023; 18:485-491. [DOI: 10.4103/1673-5374.346484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
20
|
Shibly AZ, Sheikh AM, Michikawa M, Tabassum S, Azad AK, Zhou X, Zhang Y, Yano S, Nagai A. Analysis of Cerebral Small Vessel Changes in AD Model Mice. Biomedicines 2022; 11:50. [PMID: 36672558 PMCID: PMC9855388 DOI: 10.3390/biomedicines11010050] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Amyloid β (Aβ) peptide is deposited in the brains of sporadic Alzheimer's disease (AD) due to impaired vessel-dependent clearance. To understand the mechanisms, we investigated time-dependent cerebrovascular changes in AD model mice. Cerebrovascular and other pathological changes were analyzed in AD model mice (J20 strain) aging from 2 to 9 months by immunostaining. At 2 months, Aβ was only intraneuronal, whereas vessels were positive from 3 months in J20 mice. Compared to wild-type (WT), vessel density was increased at 2 months but decreased at 9 months in J20 mice, claudin-5 levels were decreased, and vascular endothelial growth factor (VEGF) levels were increased in the cortex and hippocampus of J20 mice brain at all time points. Albumin extravasation was evident from 3 months in J20 brains. Collagen 4 was increased at 2 and 3 months. Aquaporin 4 was spread beyond the vessels starting from 3 months in J20, which was restricted around the vessel in wild-type mice. In conclusion, the study showed that an early decrease in claudin-5 was associated with VEGF expression, indicating dysfunction of the blood-brain barrier. Decreased claudin-5 might cause the leakage of blood constituents into the parenchyma that alters astrocyte polarity and its functions.
Collapse
Affiliation(s)
- Abu Zaffar Shibly
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.Z.S.); (A.K.A.); (X.Z.); (Y.Z.)
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Abdullah Md. Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Abul Kalam Azad
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.Z.S.); (A.K.A.); (X.Z.); (Y.Z.)
| | - Xiaojing Zhou
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.Z.S.); (A.K.A.); (X.Z.); (Y.Z.)
| | - Yuchi Zhang
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.Z.S.); (A.K.A.); (X.Z.); (Y.Z.)
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.M.S.); (S.T.); (S.Y.)
| | - Atsushi Nagai
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan; (A.Z.S.); (A.K.A.); (X.Z.); (Y.Z.)
| |
Collapse
|
21
|
Rustenhoven J, Kipnis J. Brain borders at the central stage of neuroimmunology. Nature 2022; 612:417-429. [PMID: 36517712 PMCID: PMC10205171 DOI: 10.1038/s41586-022-05474-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/24/2022] [Indexed: 12/16/2022]
Abstract
The concept of immune privilege suggests that the central nervous system is isolated from the immune system. However, recent studies have highlighted the borders of the central nervous system as central sites of neuro-immune interactions. Although the nervous and immune systems both function to maintain homeostasis, under rare circumstances, they can develop pathological interactions that lead to neurological or psychiatric diseases. Here we discuss recent findings that dissect the key anatomical, cellular and molecular mechanisms that enable neuro-immune responses at the borders of the brain and spinal cord and the implications of these interactions for diseases of the central nervous system.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Center for Brain immunology and Glia (BIG), Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand.
- Center for Brain Research, The University of Auckland, Auckland, New Zealand.
| | - Jonathan Kipnis
- Center for Brain immunology and Glia (BIG), Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
| |
Collapse
|
22
|
Ali AM, Alameri RA, Hendawy AO, Al-Amer R, Shahrour G, Ali EM, Alkhamees AA, Ibrahim N, Hassan BH. Psychometric evaluation of the depression anxiety stress scale 8-items (DASS-8)/DASS-12/DASS-21 among family caregivers of patients with dementia. Front Public Health 2022; 10:1012311. [PMID: 36388286 PMCID: PMC9641276 DOI: 10.3389/fpubh.2022.1012311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/26/2022] [Indexed: 01/27/2023] Open
Abstract
Patients with dementia express a set of problematic and deteriorating symptoms, along with self-care dependency. Over time, the mental health of family caregivers of persons with dementia may be affected, putting them at a high risk for psychopathology, which may be associated with endangered wellbeing of people with dementia. This cross-sectional instrumental design study examined the psychometric properties of the Depression Anxiety Stress Scale 8-items (DASS-8), DASS-12, and DASS-21 in a convenient sample of 571 caregivers from northern Italy and southern Switzerland (mean age = 53 years, SD = 12, range = 24-89 years). A bifactor structure of the three measures had the best fit; some items of the DASS-12/DASS-21 failed to load on their domain-specific factors. The three-factor structure was invariant across various groups (e.g., gender and education), expressed adequate reliability and convergent validity, and had strong positive correlation with the three-item UCLA Loneliness Scale (UCLALS3). Distress scores did not differ among carers of different types of dementia (Alzheimer's disease vs. other types, e.g., vascular dementia). However, distress scores were significantly high among female individuals, adult children caregivers, those caring for dependent patients, and those who received help with care. For 54.9 and 38.8% of the latter, care was provided by relatives and health professionals, respectively. Since the DASS-8 expresses adequate psychometrics comparable with the DASS-21, it may be used as a brief measure of distress in this population.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria, Egypt
| | - Rana Ali Alameri
- Fundamentals of Nursing Department, College of Nursing, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Amin Omar Hendawy
- Department of Animal and Poultry Production, Faculty of Agriculture, Damanhour University, Damanhour, Egypt
| | - Rasmieh Al-Amer
- Faculty of Nursing, Isra University, Amman, Jordan
- School of Nursing and Midwifery, Western Sydney University, Penrith, NSW, Australia
| | - Ghada Shahrour
- Jordan University of Science and Technology, Faculty of Nursing, Irbid, Jordan
| | - Esraa M. Ali
- Department of Basic and Educational Sciences, Faculty of Education for Early Childhood, Alexandria University, Alexandria, Egypt
| | - Abdulmajeed A. Alkhamees
- Department of Medicine, Unayzah College of Medicine and Medical Sciences, Qassim University, Unayzah, Saudi Arabia
| | - Nashwa Ibrahim
- Psychiatric and Mental Health Nursing Department, Faculty of Nursing, Mansoura University, Mansoura, Egypt
| | - Bothaina Hussein Hassan
- Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Gerontological Nursing, Faculty of Nursing, Alexandria University, Alexandria, Egypt
| |
Collapse
|
23
|
Aryal M, Azadian MM, Hart AR, Macedo N, Zhou Q, Rosenthal EL, Airan RD. Noninvasive ultrasonic induction of cerebrospinal fluid flow enhances intrathecal drug delivery. J Control Release 2022; 349:434-442. [PMID: 35798095 DOI: 10.1016/j.jconrel.2022.06.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Departments of Engineering and Radiation Oncology, Loyola University Chicago, Chicago, IL, United States
| | - Matine M Azadian
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Alex R Hart
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Nicholas Macedo
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Zhou
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Stanford Cancer Center, Stanford Medical Center, Stanford, CA, United States; Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Raag D Airan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Department of Materials Science and Engineering, Stanford University School of Medicine, Stanford, CA, United States; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
24
|
Gil-Garcia CA, Alvarez EF, Garcia RC, Mendoza-Lopez AC, Gonzalez-Hermosillo LM, Garcia-Blanco MDC, Valadez ER. Essential topics about the imaging diagnosis and treatment of Hemorrhagic Stroke: a comprehensive review of the 2022 AHA guidelines. Curr Probl Cardiol 2022; 47:101328. [PMID: 35870549 DOI: 10.1016/j.cpcardiol.2022.101328] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 11/03/2022]
Abstract
Intracerebral hemorrhage (ICH) is a severe stroke with a high death rate (40 % mortality). The prevalence of hemorrhagic stroke has increased globally, with changes in the underlying cause over time as anticoagulant use and hypertension treatment have improved. The fundamental etiology of ICH and the mechanisms of harm from ICH, particularly the complex interaction between edema, inflammation, and blood product toxicity, have been thoroughly revised by the American Heart Association (AHA) in 2022. Although numerous trials have investigated the best medicinal and surgical management of ICH, there is still no discernible improvement in survival and functional tests. Small vessel diseases, such as cerebral amyloid angiopathy (CAA) or deep perforator arteriopathy (hypertensive arteriopathy), are the most common causes of spontaneous non-traumatic intracerebral hemorrhage (ICH). Even though ICH only causes 10-15% of all strokes, it contributes significantly to morbidity and mortality, with few acute or preventive treatments proven effective. Current AHA guidelines acknowledge up to 89% sensitivity for unenhanced brain CT and 81% for brain MRI. The imaging findings of both methods are helpful for initial diagnosis and follow-up, sometimes necessary a few hours after admission, especially for detecting hemorrhagic transformation or hematoma expansion. This review summarized the essential topics on hemorrhagic stroke epidemiology, risk factors, physiopathology, mechanisms of injury, current management approaches, findings in neuroimaging, goals and outcomes, recommendations for lifestyle modifications, and future research directions ICH. A list of updated references is included for each topic.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ernesto-Roldan Valadez
- Directorado de investigación, Hospital General de Mexico "Dr. Eduardo Liceaga," 06720, CDMX, Mexico; I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Radiology, 119992, Moscow, Russia.
| |
Collapse
|
25
|
Marazuela P, Paez-Montserrat B, Bonaterra-Pastra A, Solé M, Hernández-Guillamon M. Impact of Cerebral Amyloid Angiopathy in Two Transgenic Mouse Models of Cerebral β-Amyloidosis: A Neuropathological Study. Int J Mol Sci 2022; 23:ijms23094972. [PMID: 35563362 PMCID: PMC9103818 DOI: 10.3390/ijms23094972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
The pathological accumulation of parenchymal and vascular amyloid-beta (Aβ) are the main hallmarks of Alzheimer’s disease (AD) and Cerebral Amyloid Angiopathy (CAA), respectively. Emerging evidence raises an important contribution of vascular dysfunction in AD pathology that could partially explain the failure of anti-Aβ therapies in this field. Transgenic mice models of cerebral β-amyloidosis are essential to a better understanding of the mechanisms underlying amyloid accumulation in the cerebrovasculature and its interactions with neuritic plaque deposition. Here, our main objective was to evaluate the progression of both parenchymal and vascular deposition in APP23 and 5xFAD transgenic mice in relation to age and sex. We first showed a significant age-dependent accumulation of extracellular Aβ deposits in both transgenic models, with a greater increase in APP23 females. We confirmed that CAA pathology was more prominent in the APP23 mice, demonstrating a higher progression of Aβ-positive vessels with age, but not linked to sex, and detecting a pronounced burden of cerebral microbleeds (cMBs) by magnetic resonance imaging (MRI). In contrast, 5xFAD mice did not present CAA, as shown by the negligible Aβ presence in cerebral vessels and the occurrence of occasional cMBs comparable to WT mice. In conclusion, the APP23 mouse model is an interesting tool to study the overlap between vascular and parenchymal Aβ deposition and to evaluate future disease-modifying therapy before its translation to the clinic.
Collapse
|
26
|
Li Y, Rusinek H, Butler T, Glodzik L, Pirraglia E, Babich J, Mozley PD, Nehmeh S, Pahlajani S, Wang X, Tanzi EB, Zhou L, Strauss S, Carare RO, Theise N, Okamura N, de Leon MJ. Decreased CSF clearance and increased brain amyloid in Alzheimer's disease. Fluids Barriers CNS 2022; 19:21. [PMID: 35287702 PMCID: PMC8919541 DOI: 10.1186/s12987-022-00318-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/21/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND In sporadic Alzheimer's disease (AD), brain amyloid-beta (Aβ) deposition is believed to be a consequence of impaired Aβ clearance, but this relationship is not well established in living humans. CSF clearance, a major feature of brain glymphatic clearance (BGC), has been shown to be abnormal in AD murine models. MRI phase contrast and intrathecally delivered contrast studies have reported reduced CSF flow in AD. Using PET and tau tracer 18F-THK5117, we previously reported that the ventricular CSF clearance of the PET tracer was reduced in AD and associated with elevated brain Aβ levels. METHODS In the present study, we use two PET tracers, 18F-THK5351 and 11C-PiB to estimate CSF clearance calculated from early dynamic PET frames in 9 normal controls and 15 AD participants. RESULTS we observed that the ventricular CSF clearance measures were correlated (r = 0.66, p < 0.01), with reductions in AD of 18 and 27%, respectively. We also replicated a significant relationship between ventricular CSF clearance (18F-THK5351) and brain Aβ load (r = - 0.64, n = 24, p < 0.01). With a larger sample size, we extended our observations to show that reduced CSF clearance is associated with reductions in cortical thickness and cognitive performance. CONCLUSIONS Overall, the findings support the hypothesis that failed CSF clearance is a feature of AD that is related to Aβ deposition and to the pathology of AD. Longitudinal studies are needed to determine whether failed CSF clearance is a predictor of progressive amyloidosis or its consequence.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA.
| | - Henry Rusinek
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Tracy Butler
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Elizabeth Pirraglia
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - John Babich
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Sadek Nehmeh
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Silky Pahlajani
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Xiuyuan Wang
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Emily B Tanzi
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Liangdong Zhou
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Sara Strauss
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Neil Theise
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Mony J de Leon
- Department of Radiology, Weill Cornell Medicine, Cornell University, Brain Health Imaging Institute, 407 East 61 Street, New York, NY, 10021, USA.
| |
Collapse
|
27
|
Perosa V, Oltmer J, Munting LP, Freeze WM, Auger CA, Scherlek AA, van der Kouwe AJ, Iglesias JE, Atzeni A, Bacskai BJ, Viswanathan A, Frosch MP, Greenberg SM, van Veluw SJ. Perivascular space dilation is associated with vascular amyloid-β accumulation in the overlying cortex. Acta Neuropathol 2022; 143:331-348. [PMID: 34928427 PMCID: PMC9047512 DOI: 10.1007/s00401-021-02393-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/10/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022]
Abstract
Perivascular spaces (PVS) are compartments surrounding cerebral blood vessels that become visible on MRI when enlarged. Enlarged PVS (EPVS) are commonly seen in patients with cerebral small vessel disease (CSVD) and have been suggested to reflect dysfunctional perivascular clearance of soluble waste products from the brain. In this study, we investigated histopathological correlates of EPVS and how they relate to vascular amyloid-β (Aβ) in cerebral amyloid angiopathy (CAA), a form of CSVD that commonly co-exists with Alzheimer's disease (AD) pathology. We used ex vivo MRI, semi-automatic segmentation and validated deep-learning-based models to quantify EPVS and associated histopathological abnormalities. Severity of MRI-visible PVS during life was significantly associated with severity of MRI-visible PVS on ex vivo MRI in formalin fixed intact hemispheres and corresponded with PVS enlargement on histopathology in the same areas. EPVS were located mainly around the white matter portion of perforating cortical arterioles and their burden was associated with CAA severity in the overlying cortex. Furthermore, we observed markedly reduced smooth muscle cells and increased vascular Aβ accumulation, extending into the WM, in individually affected vessels with an EPVS. Overall, these findings are consistent with the notion that EPVS reflect impaired outward flow along arterioles and have implications for our understanding of perivascular clearance mechanisms, which play an important role in the pathophysiology of CAA and AD.
Collapse
Affiliation(s)
- Valentina Perosa
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA.
- Department of Neurology, Otto-Von-Guericke University, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Jan Oltmer
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Leon P Munting
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Whitney M Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, The Netherlands
| | - Corinne A Auger
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
| | - Ashley A Scherlek
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Andre J van der Kouwe
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Juan Eugenio Iglesias
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Centre for Medical Image Computing, University College London, London, UK
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alessia Atzeni
- Centre for Medical Image Computing, University College London, London, UK
| | - Brian J Bacskai
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
| | - Matthew P Frosch
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, J. Philip Kistler Stroke Research Center, Cambridge Str. 175, Suite 300, Boston, MA, 02114, USA
- Massachusetts General Hospital, MassGeneral Institute for Neurodegenerative Disease, Charlestown, MA, USA
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
28
|
Hladky SB, Barrand MA. The glymphatic hypothesis: the theory and the evidence. Fluids Barriers CNS 2022; 19:9. [PMID: 35115036 PMCID: PMC8815211 DOI: 10.1186/s12987-021-00282-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
The glymphatic hypothesis proposes a mechanism for extravascular transport into and out of the brain of hydrophilic solutes unable to cross the blood-brain barrier. It suggests that there is a circulation of fluid carrying solutes inwards via periarterial routes, through the interstitium and outwards via perivenous routes. This review critically analyses the evidence surrounding the mechanisms involved in each of these stages. There is good evidence that both influx and efflux of solutes occur along periarterial routes but no evidence that the principal route of outflow is perivenous. Furthermore, periarterial inflow of fluid is unlikely to be adequate to provide the outflow that would be needed to account for solute efflux. A tenet of the hypothesis is that flow sweeps solutes through the parenchyma. However, the velocity of any possible circulatory flow within the interstitium is too small compared to diffusion to provide effective solute movement. By comparison the earlier classical hypothesis describing extravascular transport proposed fluid entry into the parenchyma across the blood-brain barrier, solute movements within the parenchyma by diffusion, and solute efflux partly by diffusion near brain surfaces and partly carried by flow along "preferred routes" including perivascular spaces, white matter tracts and subependymal spaces. It did not suggest fluid entry via periarterial routes. Evidence is still incomplete concerning the routes and fate of solutes leaving the brain. A large proportion of the solutes eliminated from the parenchyma go to lymph nodes before reaching blood but the proportions delivered directly to lymph or indirectly via CSF which then enters lymph are as yet unclear. In addition, still not understood is why and how the absence of AQP4 which is normally highly expressed on glial endfeet lining periarterial and perivenous routes reduces rates of solute elimination from the parenchyma and of solute delivery to it from remote sites of injection. Neither the glymphatic hypothesis nor the earlier classical hypothesis adequately explain how solutes and fluid move into, through and out of the brain parenchyma. Features of a more complete description are discussed. All aspects of extravascular transport require further study.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
29
|
Pons V, Rivest S. Targeting Systemic Innate Immune Cells as a Therapeutic Avenue for Alzheimer Disease. Pharmacol Rev 2022; 74:1-17. [PMID: 34987086 DOI: 10.1124/pharmrev.121.000400] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer disease (AD) is the first progressive neurodegenerative disease worldwide, and the disease is characterized by an accumulation of amyloid in the brain and neurovasculature that triggers cognitive decline and neuroinflammation. The innate immune system has a preponderant role in AD. The last decade, scientists focused their efforts on therapies aiming to modulate innate immunity. The latter is of great interest, since they participate to the inflammation and phagocytose the amyloid in the brain and blood vessels. We and others have developed pharmacological approaches to stimulate these cells using various ligands. These include toll-like receptor 4, macrophage colony stimulating factor, and more recently nucleotide-binding oligomerization domain-containing 2 receptors. This review will discuss the great potential to take advantage of the innate immune system to fight naturally against amyloid β accumulation and prevent its detrimental consequence on brain functions and its vascular system. SIGNIFICANCE STATEMENT: The focus on amyloid β removal from the perivascular space rather than targeting CNS plaque formation and clearance represents a new direction with a great potential. Small molecules able to act at the level of peripheral immunity would constitute a novel approach for tackling aberrant central nervous system biology, one of which we believe would have the potential of generating a lot of interest.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Québec City, QC G1V 4G2, Canada
| |
Collapse
|
30
|
Martikainen MV, Aakko-Saksa P, van den Broek L, Cassee FR, Carare RO, Chew S, Dinnyes A, Giugno R, Kanninen KM, Malm T, Muala A, Nedergaard M, Oudin A, Oyola P, Pfeiffer TV, Rönkkö T, Saarikoski S, Sandström T, Schins RPF, Topinka J, Yang M, Zeng X, Westerink RHS, Jalava PI. TUBE Project: Transport-Derived Ultrafines and the Brain Effects. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:311. [PMID: 35010571 PMCID: PMC8751045 DOI: 10.3390/ijerph19010311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 06/14/2023]
Abstract
The adverse effects of air pollutants on the respiratory and cardiovascular systems are unquestionable. However, in recent years, indications of effects beyond these organ systems have become more evident. Traffic-related air pollution has been linked with neurological diseases, exacerbated cognitive dysfunction, and Alzheimer's disease. However, the exact air pollutant compositions and exposure scenarios leading to these adverse health effects are not known. Although several components of air pollution may be at play, recent experimental studies point to a key role of ultrafine particles (UFPs). While the importance of UFPs has been recognized, almost nothing is known about the smallest fraction of UFPs, and only >23 nm emissions are regulated in the EU. Moreover, the role of the semivolatile fraction of the emissions has been neglected. The Transport-Derived Ultrafines and the Brain Effects (TUBE) project will increase knowledge on harmful ultrafine air pollutants, as well as semivolatile compounds related to adverse health effects. By including all the major current combustion and emission control technologies, the TUBE project aims to provide new information on the adverse health effects of current traffic, as well as information for decision makers to develop more effective emission legislation. Most importantly, the TUBE project will include adverse health effects beyond the respiratory system; TUBE will assess how air pollution affects the brain and how air pollution particles might be removed from the brain. The purpose of this report is to describe the TUBE project, its background, and its goals.
Collapse
Affiliation(s)
- Maria-Viola Martikainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (M.Y.); (P.I.J.)
| | - Päivi Aakko-Saksa
- VTT Technical Research Centre of Finland Ltd., 02044 Espoo, Finland;
| | | | - Flemming R. Cassee
- Centre for Sustainability, Environment and Health, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, The Netherlands;
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3508 TD Utrecht, The Netherlands;
| | - Roxana O. Carare
- Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK;
| | - Sweelin Chew
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (S.C.); (K.M.K.); (T.M.)
| | | | - Rosalba Giugno
- Computer Science Department, University of Verona, 37129 Verona, Italy;
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (S.C.); (K.M.K.); (T.M.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland; (S.C.); (K.M.K.); (T.M.)
| | - Ala Muala
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, 901 87 Umea, Sweden; (A.M.); (A.O.); (T.S.)
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Anna Oudin
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, 901 87 Umea, Sweden; (A.M.); (A.O.); (T.S.)
| | - Pedro Oyola
- Centro Mario Molina Chile, Strategic Studies Department, Santiago 602, Chile;
| | | | - Topi Rönkkö
- Aerosol Physics Laboratory, Physics Unit, Faculty of Engineering and Natural Sciences, Tampere University, 33720 Tampere, Finland;
| | - Sanna Saarikoski
- Atmospheric Composition Research, Finnish Meteorological Institute, 00101 Helsinki, Finland;
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, 901 87 Umea, Sweden; (A.M.); (A.O.); (T.S.)
| | - Roel P. F. Schins
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Dusseldorf, Germany;
| | - Jan Topinka
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Videnska 1083, 142 20 Prague, Czech Republic;
| | - Mo Yang
- Department of Environmental and Biological Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (M.Y.); (P.I.J.)
| | - Xiaowen Zeng
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
| | - Remco H. S. Westerink
- Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3508 TD Utrecht, The Netherlands;
| | - Pasi I. Jalava
- Department of Environmental and Biological Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (M.Y.); (P.I.J.)
| |
Collapse
|
31
|
Wang HL, Zhang CL, Qiu YM, Chen AQ, Li YN, Hu B. Dysfunction of the Blood-brain Barrier in Cerebral Microbleeds: from Bedside to Bench. Aging Dis 2021; 12:1898-1919. [PMID: 34881076 PMCID: PMC8612614 DOI: 10.14336/ad.2021.0514] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Cerebral microbleeds (CMBs) are a disorder of cerebral microvessels that are characterized as small (<10 mm), hypointense, round or ovoid lesions seen on T2*-weighted gradient echo MRI. There is a high prevalence of CMBs in community-dwelling healthy older people. An increasing number of studies have demonstrated the significance of CMBs in stroke, dementia, Parkinson's disease, gait disturbances and late-life depression. Blood-brain barrier (BBB) dysfunction is considered to be the event that initializes CMBs development. However, the pathogenesis of CMBs has not yet been clearly elucidated. In this review, we introduce the pathogenesis of CMBs, hypertensive vasculopathy and cerebral amyloid angiopathy, and review recent research that has advanced our understanding of the mechanisms underlying BBB dysfunction and CMBs presence. CMBs-associated risk factors can exacerbate BBB breakdown through the vulnerability of BBB anatomical and functional changes. Finally, we discuss potential pharmacological approaches to target the BBB as therapy for CMBs.
Collapse
Affiliation(s)
| | | | | | - An-qi Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ya-nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
32
|
Sankowski R, Ahmari J, Mezö C, Hrabě de Angelis AL, Fuchs V, Utermöhlen O, Buch T, Blank T, Gomez de Agüero M, Macpherson AJ, Erny D. Commensal microbiota divergently affect myeloid subsets in the mammalian central nervous system during homeostasis and disease. EMBO J 2021; 40:e108605. [PMID: 34622466 PMCID: PMC8634130 DOI: 10.15252/embj.2021108605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 12/29/2022] Open
Abstract
The immune cells of the central nervous system (CNS) comprise parenchymal microglia and at the CNS border regions meningeal, perivascular, and choroid plexus macrophages (collectively called CNS-associated macrophages, CAMs). While previous work has shown that microglial properties depend on environmental signals from the commensal microbiota, the effects of microbiota on CAMs are unknown. By combining several microbiota manipulation approaches, genetic mouse models, and single-cell RNA-sequencing, we have characterized CNS myeloid cell composition and function. Under steady-state conditions, the transcriptional profiles and numbers of choroid plexus macrophages were found to be tightly regulated by complex microbiota. In contrast, perivascular and meningeal macrophages were affected to a lesser extent. An acute perturbation through viral infection evoked an attenuated immune response of all CAMs in germ-free mice. We further assessed CAMs in a more chronic pathological state in 5xFAD mice, a model for Alzheimer's disease, and found enhanced amyloid beta uptake exclusively by perivascular macrophages in germ-free 5xFAD mice. Our results aid the understanding of distinct microbiota-CNS macrophage interactions during homeostasis and disease, which could potentially be targeted therapeutically.
Collapse
Affiliation(s)
- Roman Sankowski
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Berta‐Ottenstein‐ProgrammeFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Jasmin Ahmari
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Charlotte Mezö
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | | | - Vidmante Fuchs
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Olaf Utermöhlen
- Institute for Medical Microbiology, Immunology and Hygiene & Center for Molecular Medicine Cologne (CMMC)University of CologneKoelnGermany
| | - Thorsten Buch
- Institute of Laboratory Animal ScienceUniversity of ZurichZurichSwitzerland
| | - Thomas Blank
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Mercedes Gomez de Agüero
- Maurice E. Müller LaboratoriesDepartment for Biomedical Research (DBMR)University Clinic of Visceral Surgery and MedicineInselspitalUniversity of BernBernSwitzerland
| | - Andrew J Macpherson
- Maurice E. Müller LaboratoriesDepartment for Biomedical Research (DBMR)University Clinic of Visceral Surgery and MedicineInselspitalUniversity of BernBernSwitzerland
| | - Daniel Erny
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Berta‐Ottenstein‐Programme for Advanced Clinician ScientistsFaculty of MedicineUniversity of FreiburgFreiburgGermany
| |
Collapse
|
33
|
Balzano RF, Mannatrizio D, Castorani G, Perri M, Pennelli AM, Izzo R, Popolizio T, Guglielmi G. Imaging of Cerebral Microbleeds: Primary Patterns and Differential Diagnosis. CURRENT RADIOLOGY REPORTS 2021. [DOI: 10.1007/s40134-021-00390-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
34
|
Matsuzono K, Suzuki M, Miura K, Ozawa T, Mashiko T, Koide R, Tanaka R, Fujimoto S. Internal Jugular Vein Velocity and Spontaneous Echo Contrast Correlate with Alzheimer's Disease and Cognitive Function. J Alzheimers Dis 2021; 84:787-796. [PMID: 34602471 DOI: 10.3233/jad-210490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Many issues persist in the today's Alzheimer's disease (AD) screening and the breakthrough method is desired. OBJECTIVE We aim to validate the association between venous reflux and AD, and to develop a new method for AD screening. METHODS We examined spontaneous echo contrast, area, diameter, retrograde velocity, and anterograde velocity of the bilateral cervical internal jugular vein (IJV) using carotid ultrasonography. RESULTS A total of 112 patients participated in this study, with 26 diagnosed as AD. The proportion of both or either IJV spontaneous echo contrast (+) occupied 25 of total 26 AD patients, which showed 96.2%of sensitivity and 98.5%negative predictive value. The IJV velocities also showed significant correlation with AD diagnosis, although the IJV area or diameter did not. CONCLUSION Our results indicate that the validation of the spontaneous echo contrast or velocities of the IJV are convenient AD diagnosis screening methods and that the venous reflux disturbance correlates with AD development.
Collapse
Affiliation(s)
- Kosuke Matsuzono
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Masayuki Suzuki
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Kumiko Miura
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Tadashi Ozawa
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Takafumi Mashiko
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Reiji Koide
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Ryota Tanaka
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| | - Shigeru Fujimoto
- Division of Neurology, Department of Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
35
|
Cerebrovascular alterations in NAFLD: Is it increasing our risk of Alzheimer's disease? Anal Biochem 2021; 636:114387. [PMID: 34537182 DOI: 10.1016/j.ab.2021.114387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/27/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.
Collapse
|
36
|
Szu JI, Obenaus A. Cerebrovascular phenotypes in mouse models of Alzheimer's disease. J Cereb Blood Flow Metab 2021; 41:1821-1841. [PMID: 33557692 PMCID: PMC8327123 DOI: 10.1177/0271678x21992462] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurological degenerative disorder and is the most common cause of dementia in the elderly. Clinically, AD manifests with memory and cognitive decline associated with deposition of hallmark amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Although the mechanisms underlying AD remains unclear, two hypotheses have been proposed. The established amyloid hypothesis states that Aβ accumulation is the basis of AD and leads to formation of NFTs. In contrast, the two-hit vascular hypothesis suggests that early vascular damage leads to increased accumulation of Aβ deposits in the brain. Multiple studies have reported significant morphological changes of the cerebrovasculature which can result in severe functional deficits. In this review, we delve into known structural and functional vascular alterations in various mouse models of AD and the cellular and molecular constituents that influence these changes to further disease progression. Many studies shed light on the direct impact of Aβ on the cerebrovasculature and how it is disrupted during the progression of AD. However, more research directed towards an improved understanding of how the cerebrovasculature is modified over the time course of AD is needed prior to developing future interventional strategies.
Collapse
Affiliation(s)
- Jenny I Szu
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - André Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
37
|
Kim HJ, Cho H, Park M, Kim JW, Ahn SJ, Lyoo CH, Suh SH, Ryu YH. MRI-Visible Perivascular Spaces in the Centrum Semiovale Are Associated with Brain Amyloid Deposition in Patients with Alzheimer Disease-Related Cognitive Impairment. AJNR Am J Neuroradiol 2021; 42:1231-1238. [PMID: 33985952 DOI: 10.3174/ajnr.a7155] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 01/21/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE The association of perivascular spaces in the centrum semiovale with amyloid accumulation among patients with Alzheimer disease-related cognitive impairment is unknown. We evaluated this association in patients with Alzheimer disease-related cognitive impairment and β-amyloid deposition, assessed with [18F] florbetaben PET/CT. MATERIALS AND METHODS MR imaging and [18F] florbetaben PET/CT images of 144 patients with Alzheimer disease-related cognitive impairment were retrospectively evaluated. MR imaging-visible perivascular spaces were rated on a 4-point visual scale: a score of ≥3 or <3 indicated a high or low degree of MR imaging-visible perivascular spaces, respectively. Amyloid deposition was evaluated using the brain β-amyloid plaque load scoring system. RESULTS Compared with patients negative for β-amyloid, those positive for it were older and more likely to have lower cognitive function, a diagnosis of Alzheimer disease, white matter hyperintensity, the Apolipoprotein E ε4 allele, and a high degree of MR imaging-visible perivascular spaces in the centrum semiovale. Multivariable analysis, adjusted for age and Apolipoprotein E status, revealed that a high degree of MR imaging-visible perivascular spaces in the centrum semiovale was independently associated with β-amyloid positivity (odds ratio, 2.307; 95% CI, 1.036-5.136; P = .041). CONCLUSIONS A high degree of MR imaging-visible perivascular spaces in the centrum semiovale independently predicted β-amyloid positivity in patients with Alzheimer disease-related cognitive impairment. Thus, MR imaging-visible perivascular spaces in the centrum semiovale are associated with amyloid pathology of the brain and could be an indirect imaging marker of amyloid burden in patients with Alzheimer disease-related cognitive impairment.
Collapse
Affiliation(s)
- H J Kim
- From the Department of Nuclear Medicine (H.J.K., Y.H.R.)
- Department of Nuclear Medicine (H.J.K.), Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, South Korea
| | | | - M Park
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - J W Kim
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - S J Ahn
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | | | - S H Suh
- Radiology (M.P., J.W.K., S.J.A., S.H.S.), Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Y H Ryu
- From the Department of Nuclear Medicine (H.J.K., Y.H.R.)
| |
Collapse
|
38
|
Schmid F, Conti G, Jenny P, Weber B. The severity of microstrokes depends on local vascular topology and baseline perfusion. eLife 2021; 10:60208. [PMID: 34003107 PMCID: PMC8421069 DOI: 10.7554/elife.60208] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 05/17/2021] [Indexed: 01/26/2023] Open
Abstract
Cortical microinfarcts are linked to pathologies like cerebral amyloid angiopathy and dementia. Despite their relevance for disease progression, microinfarcts often remain undetected and the smallest scale of blood flow disturbance has not yet been identified. We employed blood flow simulations in realistic microvascular networks from the mouse cortex to quantify the impact of single-capillary occlusions. Our simulations reveal that the severity of a microstroke is strongly affected by the local vascular topology and the baseline flow rate in the occluded capillary. The largest changes in perfusion are observed in capillaries with two inflows and two outflows. This specific topological configuration only occurs with a frequency of 8%. The majority of capillaries have one inflow and one outflow and is likely designed to efficiently supply oxygen and nutrients. Taken together, microstrokes bear potential to induce a cascade of local disturbances in the surrounding tissue, which might accumulate and impair energy supply locally.
Collapse
Affiliation(s)
- Franca Schmid
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Giulia Conti
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Patrick Jenny
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Zhang X, O’Callaghan P, Li H, Tan Y, Zhang G, Barash U, Wang X, Lannfelt L, Vlodavsky I, Lindahl U, Li JP. Heparanase overexpression impedes perivascular clearance of amyloid-β from murine brain: relevance to Alzheimer's disease. Acta Neuropathol Commun 2021; 9:84. [PMID: 33971986 PMCID: PMC8111754 DOI: 10.1186/s40478-021-01182-x] [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: 03/03/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Defective amyloid-β (Aβ) clearance from the brain is a major contributing factor to the pathophysiology of Alzheimer's disease (AD). Aβ clearance is mediated by macrophages, enzymatic degradation, perivascular drainage along the vascular basement membrane (VBM) and transcytosis across the blood-brain barrier (BBB). AD pathology is typically associated with cerebral amyloid angiopathy due to perivascular accumulation of Aβ. Heparan sulfate (HS) is an important component of the VBM, thought to fulfill multiple roles in AD pathology. We previously showed that macrophage-mediated clearance of intracortically injected Aβ was impaired in the brains of transgenic mice overexpressing heparanase (Hpa-tg). This study revealed that perivascular drainage was impeded in the Hpa-tg brain, evidenced by perivascular accumulation of the injected Aβ in the thalamus of Hpa-tg mice. Furthermore, endogenous Aβ accumulated at the perivasculature of Hpa-tg thalamus, but not in control thalamus. This perivascular clearance defect was confirmed following intracortical injection of dextran that was largely retained in the perivasculature of Hpa-tg brains, compared to control brains. Hpa-tg brains presented with thicker VBMs and swollen perivascular astrocyte endfeet, as well as elevated expression of the BBB-associated water-pump protein aquaporin 4 (AQP4). Elevated levels of both heparanase and AQP4 were also detected in human AD brain. These findings indicate that elevated heparanase levels alter the organization and composition of the BBB, likely through increased fragmentation of BBB-associated HS, resulting in defective perivascular drainage. This defect contributes to perivascular accumulation of Aβ in the Hpa-tg brain, highlighting a potential role for heparanase in the pathogenesis of AD.
Collapse
|
40
|
Pyun JM, Kang MJ, Ryoo N, Suh J, Youn YC, Park YH, Kim S. Amyloid Metabolism and Amyloid-Targeting Blood-Based Biomarkers of Alzheimer's Disease. J Alzheimers Dis 2021; 75:685-696. [PMID: 32390633 DOI: 10.3233/jad-200104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Amyloid-β (Aβ) is a key protein in Alzheimer's disease (AD) in that its accumulation induces complex pathological changes. Although there has been extensive research on the metabolism of Aβ in AD, new compelling results have recently emerged. Historically, the production and clearance of Aβ have been thought to originate in the central nervous system (CNS). However, recent evidence suggests that the production and clearance of Aβ can also occur in the peripheral system, and that the peripherally driven Aβ migrates to the CNS and induces amyloidopathy with subsequent AD pathologic changes in the brain. This concept implies that AD is not restricted to the CNS but is a systemic disease instead. As such, the development of blood-based biomarkers targeting Aβ is of great interest. Central and peripheral Aβ are both active contributors to the pathology of AD and interact bidirectionally. Measuring peripheral Aβ is not just observing the reflection of the residual Aβ removed from the CNS but also tracking the ongoing process of AD pathology. Additionally, blood-based biomarkers could be a more accessible tool in clinical and research settings. Through arduous research, several blood-based biomarker assays have demonstrated notable results. In this review, we describe the metabolism of Aβ and the amyloid-targeting blood-based biomarkers of AD.
Collapse
Affiliation(s)
- Jung-Min Pyun
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Nayoung Ryoo
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Jeewon Suh
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Republic of Korea
| |
Collapse
|
41
|
Cheng Y, Ma X, Belfield KD, Haorah J. Biphasic Effects of Ethanol Exposure on Waste Metabolites Clearance in the CNS. Mol Neurobiol 2021; 58:3953-3967. [PMID: 33895940 DOI: 10.1007/s12035-021-02379-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
We have shown that the effects of low-dose ethanol promote the clearance of waste metabolites, such as amyloid-beta (Aβ) proteins, from the brain through the perivascular space (PVS). We demonstrated that dilative reactivity of arterial smooth muscle and endothelial cells regulate this clearance. These findings indicate the importance of blood-brain barrier (BBB) transvascular clearance of large size metabolites from the central nervous system (CNS), where the lymphatic clearance system is absent. We next examined the contrasting effects of acute low-dose and chronic moderate ethanol exposure on BBB-associated perivascular clearance. We injected a high molecular weight fluorescent dye into the interstitial space or directly into the cerebrospinal fluid (CSF). Bio-distribution of this tracer was then examined in different brain regions by multiphoton imaging and whole brain tissue section scanning. Ethanol-induced molecular/cellular mechanisms that drive the increase or decrease in movement of the fluorescent tracer were correlated to BBB integrity and arterial vessel reactivity. We found that activation of endothelial nitric oxide synthase (eNOS) under low-dose ethanol conditions with a shift to activation of inducible NOS (iNOS) under chronic high ethanol exposure conditions, which appeared to regulate these contrasting effects. We validated these observations by qualitative and quantitative investigation of eNOS, iNOS, BBB integrity, and perivascular clearance of waste metabolites. We concluded that the effects of low-dose ethanol increased the diffusive movement of waste metabolites via eNOS-derived NO, which increased the arterial endothelial-smooth muscle cell dilative reactivity without affecting BBB integrity, whereas a prolonged induction of iNOS under chronic ethanol exposure conditions caused oxidative damage of the arterial endothelial-smooth muscle layers resulting in cerebral amyloid-like angiopathy. This led to dysfunction of the BBB, dilative reactivity, and impaired waste metabolites movement from the interstitial space or subarachnoid space (SAS) through perivascular clearance.
Collapse
Affiliation(s)
- Yiming Cheng
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Xiaotang Ma
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Kevin D Belfield
- Department of Chemistry and Environmental Science, College of Science and Liberal Arts, New Jersey Institute of Technology, 323 Martin Luther King, Jr., Blvd., Newark, NJ, 07102, USA
| | - James Haorah
- Laboratory of Neurovascular Inflammation and Neurodegeneration, Department of Biomedical Engineering, Center for Injury Bio Mechanics, Materials and Medicine, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| |
Collapse
|
42
|
Tsai HH, Pasi M, Tsai LK, Huang CC, Chen YF, Lee BC, Yen RF, Gurol ME, Jeng JS. Centrum Semiovale Perivascular Space and Amyloid Deposition in Spontaneous Intracerebral Hemorrhage. Stroke 2021; 52:2356-2362. [PMID: 33874751 DOI: 10.1161/strokeaha.120.032139] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei (H.-H.T.).,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei (H.-H.T.).,Department of Neurology (H.-H.T., L.-K.T., J.-S.J.), National Taiwan University Hospital, Taipei
| | - Marco Pasi
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, France (M.P.)
| | - Li-Kai Tsai
- Department of Neurology (H.-H.T., L.-K.T., J.-S.J.), National Taiwan University Hospital, Taipei
| | - Chi-Ching Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei (C.-C.H.)
| | - Ya-Fang Chen
- Department of Medical Imaging (Y.-F.C., B.-C.L.), National Taiwan University Hospital, Taipei
| | - Bo-Ching Lee
- Department of Medical Imaging (Y.-F.C., B.-C.L.), National Taiwan University Hospital, Taipei
| | - Ruoh-Fang Yen
- Department of Nuclear Medicine (R.-F.Y.), National Taiwan University Hospital, Taipei
| | - M Edip Gurol
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (M.E.G.)
| | - Jiann-Shing Jeng
- Department of Neurology (H.-H.T., L.-K.T., J.-S.J.), National Taiwan University Hospital, Taipei
| |
Collapse
|
43
|
Marazuela P, Bonaterra-Pastra A, Faura J, Penalba A, Pizarro J, Pancorbo O, Rodríguez-Luna D, Vert C, Rovira A, Pujadas F, Freijo MM, Tur S, Martínez-Zabaleta M, Cardona Portela P, Vera R, Lebrato-Hernández L, Arenillas JF, Pérez-Sánchez S, Montaner J, Delgado P, Hernández-Guillamon M. Circulating AQP4 Levels in Patients with Cerebral Amyloid Angiopathy-Associated Intracerebral Hemorrhage. J Clin Med 2021; 10:jcm10050989. [PMID: 33801197 PMCID: PMC7957864 DOI: 10.3390/jcm10050989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/27/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a major cause of lobar intracerebral hemorrhage (ICH) in elderly patients. Growing evidence suggests a potential role of aquaporin 4 (AQP4) in amyloid-beta-associated diseases, including CAA pathology. Our aim was to investigate the circulating levels of AQP4 in a cohort of patients who had suffered a lobar ICH with a clinical diagnosis of CAA. AQP4 levels were analyzed in the serum of 60 CAA-related ICH patients and 19 non-stroke subjects by enzyme-linked immunosorbent assay (ELISA). The CAA–ICH cohort was divided according to the time point of the functional outcome evaluation: mid-term (12 ± 18.6 months) and long-term (38.5 ± 32.9 months) after the last ICH. Although no differences were found in AQP4 serum levels between cases and controls, lower levels were found in CAA patients presenting specific hemorrhagic features such as ≥2 lobar ICHs and ≥5 lobar microbleeds detected by magnetic resonance imaging (MRI). In addition, CAA-related ICH patients who presented a long-term good functional outcome had higher circulating AQP4 levels than subjects with a poor outcome or controls. Our data suggest that AQP4 could potentially predict a long-term functional outcome and may play a protective role after a lobar ICH.
Collapse
Affiliation(s)
- Paula Marazuela
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Júlia Faura
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Anna Penalba
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Jesús Pizarro
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Olalla Pancorbo
- Stroke Unit, Department of Neurology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (O.P.); (D.R.-L.)
| | - David Rodríguez-Luna
- Stroke Unit, Department of Neurology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (O.P.); (D.R.-L.)
| | - Carla Vert
- Neuroradiology, Department of Radiology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (C.V.); (A.R.)
| | - Alex Rovira
- Neuroradiology, Department of Radiology, Vall d’Hebron Hospital, 08035 Barcelona, Spain; (C.V.); (A.R.)
| | - Francesc Pujadas
- Dementia Unit, Neurology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain;
| | - M. Mar Freijo
- Neurovascular Group, Biocruces Health Research Institute, 48903 Barakaldo, Spain;
| | - Silvia Tur
- Neurology, Son Espases University Hospital, 07120 Balearic Islands, Spain;
| | | | - Pere Cardona Portela
- Department of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, 08907 Barcelona, Spain;
| | - Rocío Vera
- Stroke Unit, Department of Neurology, Ramon y Cajal University Hospital, 28034 Madrid, Spain;
| | | | - Juan F. Arenillas
- Stroke Program, Department of Neurology, Hospital Clínico Universitario, 47003 Valladolid, Spain;
- Clinical Neurosciences Research Group, Department of Medicine, University of Valladolid, 47003 Valladolid, Spain
| | | | - Joan Montaner
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
- Department of Neurology, Virgen Macarena University Hospital, 41009 Sevilla, Spain;
- Stroke Research Program, Institute of Biomedicine of Sevilla, IBiS, Virgen del Rocío University Hospital, University of Sevilla, 41009 Sevilla, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (P.M.); (A.B.-P.); (J.F.); (A.P.); (J.P.); (J.M.); (P.D.)
- Correspondence:
| |
Collapse
|
44
|
Liu CC, Yamazaki Y, Heckman MG, Martens YA, Jia L, Yamazaki A, Diehl NN, Zhao J, Zhao N, DeTure M, Davis MD, Felton LM, Qiao W, Li Y, Li H, Fu Y, Wang N, Wren M, Aikawa T, Holm ML, Oue H, Linares C, Allen M, Carrasquillo MM, Murray ME, Petersen RC, Ertekin-Taner N, Dickson DW, Kanekiyo T, Bu G. Tau and apolipoprotein E modulate cerebrovascular tight junction integrity independent of cerebral amyloid angiopathy in Alzheimer's disease. Alzheimers Dement 2020; 16:1372-1383. [PMID: 32827351 PMCID: PMC8103951 DOI: 10.1002/alz.12104] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 01/05/2020] [Accepted: 01/15/2020] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Cerebrovascular pathologies including cerebral amyloid angiopathy (CAA) and blood-brain barrier (BBB) dysregulation are prominent features in the majority of Alzheimer's disease (AD) cases. METHODS We performed neuropathologic and biochemical studies on a large, neuropathologically confirmed human AD cohort (N = 469). Amounts of endothelial tight junction proteins claudin-5 (CLDN5) and occludin (OCLN), and major AD-related molecules (amyloid beta [Aβ40], Aβ42, tau, p-tau, and apolipoprotein E) in the temporal cortex were assessed by ELISA. RESULTS Higher levels of soluble tau, insoluble p-tau, and apolipoprotein E (apoE) were independently correlated with lower levels of endothelial tight junction proteins CLDN5 and OCLN in AD brains. Although high Aβ40 levels, APOE ε4, and male sex were predominantly associated with exacerbated CAA severity, those factors did not influence tight junction protein levels. DISCUSSION Refining the molecular mechanisms connecting tau, Aβ, and apoE with cerebrovascular pathologies is critical for greater understanding of AD pathogenesis and establishing effective therapeutic interventions for the disease.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Michael G. Heckman
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida, USA
| | - Yuka A. Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Nancy N. Diehl
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Jacksonville, Florida, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Michael DeTure
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Mary D. Davis
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Hongmei Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Melissa Wren
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Tomonori Aikawa
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Hiroshi Oue
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | | | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
45
|
Intracisternal injection of beta-amyloid seeds promotes cerebral amyloid angiopathy. Brain Behav Immun 2020; 89:628-640. [PMID: 32739364 DOI: 10.1016/j.bbi.2020.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/19/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
Beta amyloid (Aβ) is a key component of parenchymal Aβ plaques and vascular Aβ fibrils, which lead to cerebral amyloid angiopathy (CAA) in Alzheimer's disease (AD). Recent studies have revealed that Aβ contained in the cerebrospinal fluid (CSF) can re-enter into brain through paravascular spaces. However, whether Aβ in CSF may act as a constant source of pathogenic Aβ in AD is still unclear. This study aimed to examine whether Aβ pathology could be worsened when CSF Aβ level was enhanced by intra-cisternal infusion of aged brain extract containing abundant Aβ in TgCRND8 host mice. TgCRND8 mouse is an AD animal model which develops predominant parenchymal Aβ plaques in the brain at as early as 3 months of age. Here, we showed that single intracisternal injection of Aβ seeds into TgCRND8 mice before the presence of Aβ pathology induced robust prion-like propagation of CAA within 90 days. The induced CAA is mainly distributed in the cerebral cortex, hippocampus and thalamus of TgCRND8 mice. Surprisingly, despite the robust increase in CAA levels, the TgCRND8 mice had a marked decrease in parenchymal Aβ plaques and the plaques related neuroinflammation in the brains compared with the control mice. These results amply indicate that Aβ in CSF may act as a source of Aβ contributing to the growth of vascular Aβ deposits in CAA. Our findings provide experimental evidence to unravel the mechanisms of CAA formation and the potential of targeting CSF Aβ for CAA.
Collapse
|
46
|
Ali AM, Kunugi H. Royal Jelly as an Intelligent Anti-Aging Agent-A Focus on Cognitive Aging and Alzheimer's Disease: A Review. Antioxidants (Basel) 2020; 9:E937. [PMID: 33003559 PMCID: PMC7601550 DOI: 10.3390/antiox9100937] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023] Open
Abstract
The astronomical increase of the world's aged population is associated with the increased prevalence of neurodegenerative diseases, heightened disability, and extremely high costs of care. Alzheimer's Disease (AD) is a widespread, age-related, multifactorial neurodegenerative disease that has enormous social and financial drawbacks worldwide. The unsatisfactory outcomes of available AD pharmacotherapy necessitate the search for alternative natural resources that can target various the underlying mechanisms of AD pathology and reduce disease occurrence and/or progression. Royal jelly (RJ) is the main food of bee queens; it contributes to their fertility, long lifespan, and memory performance. It represents a potent nutraceutical with various pharmacological properties, and has been used in a number of preclinical studies to target AD and age-related cognitive deterioration. To understand the mechanisms through which RJ affects cognitive performance both in natural aging and AD, we reviewed the literature, elaborating on the metabolic, molecular, and cellular mechanisms that mediate its anti-AD effects. Preclinical findings revealed that RJ acts as a multidomain cognitive enhancer that can restore cognitive performance in aged and AD models. It promotes brain cell survival and function by targeting multiple adversities in the neuronal microenvironment such as inflammation, oxidative stress, mitochondrial alterations, impaired proteostasis, amyloid-β toxicity, Ca excitotoxicity, and bioenergetic challenges. Human trials using RJ in AD are limited in quantity and quality. Here, the limitations of RJ-based treatment strategies are discussed, and directions for future studies examining the effect of RJ in cognitively impaired subjects are noted.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria 21527, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| |
Collapse
|
47
|
Kwon S, Moreno-Gonzalez I, Taylor-Presse K, Edwards Iii G, Gamez N, Calderon O, Zhu B, Velasquez FC, Soto C, Sevick-Muraca EM. Impaired Peripheral Lymphatic Function and Cerebrospinal Fluid Outflow in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 69:585-593. [PMID: 31104026 DOI: 10.3233/jad-190013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cerebrospinal fluid (CSF) outflow from the brain occurs through absorption into the arachnoid villi and, more predominantly, through meningeal and olfactory lymphatics that ultimately drain into the peripheral lymphatics. Impaired CSF outflow has been postulated as a contributing mechanism in Alzheimer's disease (AD). Herein we conducted near-infrared fluorescence imaging of CSF outflow into the peripheral lymph nodes (LNs) and of peripheral lymphatic function in a transgenic mouse model of AD (5XFAD) and wild-type (WT) littermates. CSF outflow was assessed from change in fluorescence intensity in the submandibular LNs as a function of time following bolus, an intrathecal injection of indocyanine green (ICG). Peripheral lymphatic function was measured by assessing lymphangion contractile function in lymphatics draining into the popliteal LN following intradermal ICG injection in the dorsal aspect of the hind paw. The results show 1) significantly impaired CSF outflow into the submandibular LNs of 5XFAD mice and 2) reduced contractile frequency in the peripheral lymphatics as compared to WT mice. Impaired CSF clearance was also evidenced by reduction of fluorescence on ventral surfaces of extracted brains of 5XFAD mice at euthanasia. These results support the hypothesis that lymphatic congestion caused by reduced peripheral lymphatic function could limit CSF outflow and may contribute to the cause and/or progression of AD.
Collapse
Affiliation(s)
- Sunkuk Kwon
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Ines Moreno-Gonzalez
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Kathleen Taylor-Presse
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - George Edwards Iii
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Nazaret Gamez
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Olivia Calderon
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Banghe Zhu
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Fred Christian Velasquez
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Claudio Soto
- Mitchell Center for Alzheimer's disease and Related Brain Disorders, Department of Neurology, The University of Texas Health Science Center, Houston, TX, USA
| | - Eva M Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| |
Collapse
|
48
|
Rivera-Rivera LA, Cody KA, Rutkowski D, Cary P, Eisenmenger L, Rowley HA, Carlsson CM, Johnson SC, Johnson KM. Intracranial vascular flow oscillations in Alzheimer's disease from 4D flow MRI. Neuroimage Clin 2020; 28:102379. [PMID: 32871386 PMCID: PMC7476069 DOI: 10.1016/j.nicl.2020.102379] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/10/2020] [Accepted: 08/07/2020] [Indexed: 11/26/2022]
Abstract
Recent modeling and experimental evidence suggests clearance of soluble metabolites from the brain can be driven by low frequency flow oscillations (LFOs) through the intramural periarterial drainage (IPAD) pathway. This study investigates the use of 4D flow MRI to derive LFOs from arterial and venous measures of blood flow. 3D radial 4D flow MRI data were acquired on a 3.0 T scanner and reconstructed using a low-rank constraint to produce time resolved measurements of blood flow. Physical phantom experiments were performed to validate the time resolved 4D flow against a standard 2D phase contrast (PC) approach. To evaluate the ability of 4D flow to distinguish physiologic flow changes from noise, healthy volunteers were scanned during a breath-hold (BH) maneuver and compared against 2D PC measures. Finally, flow measures were performed in intracranial arteries and veins of 112 participants including subjects diagnosed with Alzheimer's disease (AD) clinical syndrome (n = 23), and healthy controls (n = 89) on whom apolipoprotein ɛ4 positivity (APOE4+) and parental history of AD dementia (FH+) was known. To assess LFOs, flow range, standard deviation, demeaned temporal flow changes, and power spectral density were quantified from the time series. Group differences were assessed using ANOVA followed by Tukey-Kramer method for pairwise comparison for adjusted means (P < 0.05). Significantly lower LFOs as measured from flow variation range and standard deviations were observed in the arteries of AD subjects when compared to age-matched controls (P = 0.005, P = 0.011). Results suggest altered vascular function in AD subjects. 4D flow based spontaneous LFO measures might hold potential for longitudinal studies aimed at predicting cognitive trajectories in AD and study disease mechanisms.
Collapse
Affiliation(s)
- Leonardo A Rivera-Rivera
- Department of Medical Physics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Karly A Cody
- Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - David Rutkowski
- Department of Radiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Paul Cary
- Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Laura Eisenmenger
- Department of Radiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Howard A Rowley
- Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M Carlsson
- Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Kevin M Johnson
- Department of Medical Physics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
49
|
Harrison IF, Ismail O, Machhada A, Colgan N, Ohene Y, Nahavandi P, Ahmed Z, Fisher A, Meftah S, Murray TK, Ottersen OP, Nagelhus EA, O’Neill MJ, Wells JA, Lythgoe MF. Impaired glymphatic function and clearance of tau in an Alzheimer's disease model. Brain 2020; 143:2576-2593. [PMID: 32705145 PMCID: PMC7447521 DOI: 10.1093/brain/awaa179] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/28/2020] [Accepted: 04/13/2020] [Indexed: 01/09/2023] Open
Abstract
The glymphatic system, that is aquaporin 4 (AQP4) facilitated exchange of CSF with interstitial fluid (ISF), may provide a clearance pathway for protein species such as amyloid-β and tau, which accumulate in the brain in Alzheimer's disease. Further, tau protein transference via the extracellular space, the compartment that is cleared by the glymphatic pathway, allows for its neuron-to-neuron propagation, and the regional progression of tauopathy in the disorder. The glymphatic system therefore represents an exciting new target for Alzheimer's disease. Here we aim to understand the involvement of glymphatic CSF-ISF exchange in tau pathology. First, we demonstrate impaired CSF-ISF exchange and AQP4 polarization in a mouse model of tauopathy, suggesting that this clearance pathway may have the potential to exacerbate or even induce pathogenic accumulation of tau. Subsequently, we establish the central role of AQP4 in the glymphatic clearance of tau from the brain; showing marked impaired glymphatic CSF-ISF exchange and tau protein clearance using the novel AQP4 inhibitor, TGN-020. As such, we show that this system presents as a novel druggable target for the treatment of Alzheimer's disease, and possibly other neurodegenerative diseases alike.
Collapse
Affiliation(s)
- Ian F Harrison
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| | - Ozama Ismail
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| | - Asif Machhada
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Niall Colgan
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
- School of Physics, National University of Ireland Galway, Ireland
| | - Yolanda Ohene
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| | - Payam Nahavandi
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| | - Zeshan Ahmed
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, UK
| | - Alice Fisher
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, UK
| | - Soraya Meftah
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, UK
| | - Tracey K Murray
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey, UK
| | - Ole P Ottersen
- Office of the President, Karolinska Institutet, Stockholm, Sweden
| | - Erlend A Nagelhus
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Jack A Wells
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Department of Imaging, Division of Medicine, University College London, London, UK
| |
Collapse
|
50
|
Nimmo J, Johnston DA, Dodart JC, MacGregor-Sharp MT, Weller RO, Nicoll JAR, Verma A, Carare RO. Peri-arterial pathways for clearance of α-Synuclein and tau from the brain: Implications for the pathogenesis of dementias and for immunotherapy. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2020; 12:e12070. [PMID: 32782922 PMCID: PMC7409108 DOI: 10.1002/dad2.12070] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
Introduction Accumulation of amyloid beta (Aβ), α-synuclein (αSyn), and tau in dementias indicates their age-related failure of elimination from the brain. Aβ is eliminated along basement membranes in walls of cerebral arterioles and leptomeningeal arteries (intramural peri-arterial drainage [IPAD]); IPAD is impaired with age. We test the hypothesis that αSyn and tau are also eliminated from the normal brain along IPAD pathways. Methods Soluble αSyn or tau was injected into mouse hippocampus. Animals were perfused 5 minutes to 7 days post-injection. Blood vessels were identified by ROX-SE for light-sheet and immunolabeling for confocal microscopy. IPAD was quantified by measuring the proportion of arterioles with αSyn/tau. Results αSyn and tau are eliminated from the brain by IPAD but with different dynamics. Discussion Age-related failure of IPAD may play a role in the pathogenesis of synucleinopathies and tauopathies. αSyn persists within IPAD at 24 hours, which may affect immunotherapy for αSyn.
Collapse
Affiliation(s)
- Jacqui Nimmo
- Faculty of Medicine University of Southampton Southampton UK
| | | | - J C Dodart
- United Neuroscience Dublin Republic of Ireland
| | | | - Roy O Weller
- Faculty of Medicine University of Southampton Southampton UK
| | | | - Ajay Verma
- United Neuroscience Dublin Republic of Ireland
| | - Roxana O Carare
- Faculty of Medicine University of Southampton Southampton UK
| |
Collapse
|