1
|
Pramotton FM, Spitz S, Kamm RD. Challenges and Future Perspectives in Modeling Neurodegenerative Diseases Using Organ-on-a-Chip Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403892. [PMID: 38922799 PMCID: PMC11348103 DOI: 10.1002/advs.202403892] [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: 04/12/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Neurodegenerative diseases (NDDs) affect more than 50 million people worldwide, posing a significant global health challenge as well as a high socioeconomic burden. With aging constituting one of the main risk factors for some NDDs such as Alzheimer's disease (AD) and Parkinson's disease (PD), this societal toll is expected to rise considering the predicted increase in the aging population as well as the limited progress in the development of effective therapeutics. To address the high failure rates in clinical trials, legislative changes permitting the use of alternatives to traditional pre-clinical in vivo models are implemented. In this regard, microphysiological systems (MPS) such as organ-on-a-chip (OoC) platforms constitute a promising tool, due to their ability to mimic complex and human-specific tissue niches in vitro. This review summarizes the current progress in modeling NDDs using OoC technology and discusses five critical aspects still insufficiently addressed in OoC models to date. Taking these aspects into consideration in the future MPS will advance the modeling of NDDs in vitro and increase their translational value in the clinical setting.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
2
|
Munsterman D, Falcione S, Long R, Boghozian R, Joy T, Camicioli R, Smith EE, Jickling GC. Cerebral amyloid angiopathy and the immune system. Alzheimers Dement 2024; 20:4999-5008. [PMID: 38881491 PMCID: PMC11247707 DOI: 10.1002/alz.13826] [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: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 06/18/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of amyloid protein in the walls of cerebral blood vessels. This deposition of amyloid causes damage to the cerebral vasculature, resulting in blood-brain barrier disruption, cerebral hemorrhage, cognitive decline, and dementia. The role of the immune system in CAA is complex and not fully understood. While the immune system has a clear role in the rare inflammatory variants of CAA (CAA related inflammation and Abeta related angiitis), the more common variants of CAA also have immune system involvement. In a protective role, immune cells may facilitate the clearance of beta-amyloid from the cerebral vasculature. The immune system can also contribute to CAA pathology, promoting vascular injury, blood-brain barrier breakdown, inflammation, and progression of CAA. In this review, we summarize the role of the immune system in CAA, including the potential of immune based treatment strategies to slow vascular disease in CAA and associated cognitive impairment, white matter disease progression, and reduce the risk of cerebral hemorrhage. HIGHLIGHTS: The immune system has a role in cerebral amyloid angiopathy (CAA) which is summarized in this review. There is an inflammatory response to beta-amyloid that may contribute to brain injury and cognitive impairment. Immune cells may facilitate the clearance of beta-amyloid from the cerebral vasculature. Improved understanding of the immune system in CAA may afford novel treatment to improve outcomes in patients with CAA.
Collapse
Affiliation(s)
| | - Sarina Falcione
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | - Rebecca Long
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Twinkle Joy
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Eric E. Smith
- Clinical NeurosciencesHotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | | |
Collapse
|
3
|
Rothenberg KG, Bekris L, Leverenz JB, Wu J, Lee J, Statsevych V, Ruggieri P, Jones SE. Cerebral Amyloid Angiopathy in Patients with Cognitive Impairment: Cerebrospinal Fluid Biomarkers. Dement Geriatr Cogn Disord 2024; 53:248-254. [PMID: 38889704 PMCID: PMC11446477 DOI: 10.1159/000539884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
INTRODUCTION Cerebral amyloid angiopathy (CAA) is characterized by amyloid β (Aβ) deposition in brain vessels, leading to hemorrhagic phenomena and cognitive impairment. Magnetic resonance imaging (MRI)-based criteria allow a diagnosis of probable CAA in vivo, but such a diagnosis cannot predict the eventual development of CAA. METHODS We conducted a retrospective cohort study of 464 patients with cognitive disorders whose data were included in a brain health biobank. De-identified parameters including sex, age, cognitive score, APOE status, and cerebrospinal fluid (CSF) levels of Aβ 1-40, Aβ 1-42, phosphorylated tau, and total tau were assessed in those with and without CAA. Odds ratios (ORs) and 95% confidence intervals (CIs) were determined. RESULTS CAA was present in 53 of 464 (11.5%) patients. P-tau level was significantly higher in those with CAA (115 vs. 84.3 pg/mL p = 0.038). In univariate analyses, the risk of developing CAA was higher with increased age (OR, 1.036; 95% CI: 1.008, 1.064; p = 0.011) and decreased CSF level of Aβ 1-40 (OR, 0.685; 95% CI: 0.534, 0.878; p = 0.003). In multivariate analyses, the risk of CAA remained higher with a decreased CSF level of Aβ 1-40 (OR, 0.681; 95% CI: 0.531, 0.874; p = 0.003). CONCLUSION These findings suggest that Aβ 1-40 levels in the CSF might be a useful molecular biomarker of CAA in patients with dementia.
Collapse
Affiliation(s)
- Kasia Gustaw Rothenberg
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Lynn Bekris
- Genomic Medicine Institute Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jenny Wu
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jonathan Lee
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Paul Ruggieri
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stephen E Jones
- Department of Radiology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Bhattarai P, Gunasekaran TI, Belloy ME, Reyes-Dumeyer D, Jülich D, Tayran H, Yilmaz E, Flaherty D, Turgutalp B, Sukumar G, Alba C, McGrath EM, Hupalo DN, Bacikova D, Le Guen Y, Lantigua R, Medrano M, Rivera D, Recio P, Nuriel T, Ertekin-Taner N, Teich AF, Dickson DW, Holley S, Greicius M, Dalgard CL, Zody M, Mayeux R, Kizil C, Vardarajan BN. Rare genetic variation in fibronectin 1 (FN1) protects against APOEε4 in Alzheimer's disease. Acta Neuropathol 2024; 147:70. [PMID: 38598053 PMCID: PMC11006751 DOI: 10.1007/s00401-024-02721-1] [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: 12/11/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
The risk of developing Alzheimer's disease (AD) significantly increases in individuals carrying the APOEε4 allele. Elderly cognitively healthy individuals with APOEε4 also exist, suggesting the presence of cellular mechanisms that counteract the pathological effects of APOEε4; however, these mechanisms are unknown. We hypothesized that APOEε4 carriers without dementia might carry genetic variations that could protect them from developing APOEε4-mediated AD pathology. To test this, we leveraged whole-genome sequencing (WGS) data in the National Institute on Aging Alzheimer's Disease Family Based Study (NIA-AD FBS), Washington Heights/Inwood Columbia Aging Project (WHICAP), and Estudio Familiar de Influencia Genetica en Alzheimer (EFIGA) cohorts and identified potentially protective variants segregating exclusively among unaffected APOEε4 carriers. In homozygous unaffected carriers above 70 years old, we identified 510 rare coding variants. Pathway analysis of the genes harboring these variants showed significant enrichment in extracellular matrix (ECM)-related processes, suggesting protective effects of functional modifications in ECM proteins. We prioritized two genes that were highly represented in the ECM-related gene ontology terms, (FN1) and collagen type VI alpha 2 chain (COL6A2) and are known to be expressed at the blood-brain barrier (BBB), for postmortem validation and in vivo functional studies. An independent analysis in a large cohort of 7185 APOEε4 homozygous carriers found that rs140926439 variant in FN1 was protective of AD (OR = 0.29; 95% CI [0.11, 0.78], P = 0.014) and delayed age at onset of disease by 3.37 years (95% CI [0.42, 6.32], P = 0.025). The FN1 and COL6A2 protein levels were increased at the BBB in APOEε4 carriers with AD. Brain expression of cognitively unaffected homozygous APOEε4 carriers had significantly lower FN1 deposition and less reactive gliosis compared to homozygous APOEε4 carriers with AD, suggesting that FN1 might be a downstream driver of APOEε4-mediated AD-related pathology and cognitive decline. To validate our findings, we used zebrafish models with loss-of-function (LOF) mutations in fn1b-the ortholog for human FN1. We found that fibronectin LOF reduced gliosis, enhanced gliovascular remodeling, and potentiated the microglial response, suggesting that pathological accumulation of FN1 could impair toxic protein clearance, which is ameliorated with FN1 LOF. Our study suggests that vascular deposition of FN1 is related to the pathogenicity of APOEε4, and LOF variants in FN1 may reduce APOEε4-related AD risk, providing novel clues to potential therapeutic interventions targeting the ECM to mitigate AD risk.
Collapse
Affiliation(s)
- Prabesh Bhattarai
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Tamil Iniyan Gunasekaran
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dolly Reyes-Dumeyer
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Dörthe Jülich
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA
| | - Hüseyin Tayran
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Elanur Yilmaz
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Delaney Flaherty
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Bengisu Turgutalp
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Gauthaman Sukumar
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Camille Alba
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Elisa Martinez McGrath
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Daniel N Hupalo
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Dagmar Bacikova
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Rafael Lantigua
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
- Department of Medicine, College of Physicians and Surgeons, Columbia University New York, New York, USA
| | - Martin Medrano
- School of Medicine, Pontificia Universidad Catolica Madre y Maestra, Santiago, Dominican Republic
| | - Diones Rivera
- Department of Neurology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
- School of Medicine, Universidad Pedro Henriquez Urena (UNPHU), Santo Domingo, Dominican Republic
| | - Patricia Recio
- Department of Neurology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Andrew F Teich
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Scott Holley
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, 06520, USA
| | - Michael Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- The American Genome Center, Center for Military Precision Health, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Michael Zody
- New York Genome Center, New York, NY, 10013, USA
| | - Richard Mayeux
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 W 168th St., New York, NY, 10032, USA
| | - Caghan Kizil
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA.
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Badri N Vardarajan
- Department of Neurology, Columbia University Irving Medical Center, Columbia University New York, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY, USA.
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
5
|
Cai Y, Zhang Y, Leng S, Ma Y, Jiang Q, Wen Q, Ju S, Hu J. The relationship between inflammation, impaired glymphatic system, and neurodegenerative disorders: A vicious cycle. Neurobiol Dis 2024; 192:106426. [PMID: 38331353 DOI: 10.1016/j.nbd.2024.106426] [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: 11/18/2023] [Revised: 01/16/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
The term "glymphatic" emerged roughly a decade ago, marking a pivotal point in neuroscience research. The glymphatic system, a glial-dependent perivascular network distributed throughout the brain, has since become a focal point of investigation. There is increasing evidence suggesting that impairment of the glymphatic system appears to be a common feature of neurodegenerative disorders, and this impairment exacerbates as disease progression. Nevertheless, the common factors contributing to glymphatic system dysfunction across most neurodegenerative disorders remain unclear. Inflammation, however, is suspected to play a pivotal role. Dysfunction of the glymphatic system can lead to a significant accumulation of protein and waste products, which can trigger inflammation. The interaction between the glymphatic system and inflammation appears to be cyclical and potentially synergistic. Yet, current research is limited, and there is a lack of comprehensive models explaining this association. In this perspective review, we propose a novel model suggesting that inflammation, impaired glymphatic function, and neurodegenerative disorders interconnected in a vicious cycle. By presenting experimental evidence from the existing literature, we aim to demonstrate that: (1) inflammation aggravates glymphatic system dysfunction, (2) the impaired glymphatic system exacerbated neurodegenerative disorders progression, (3) neurodegenerative disorders progression promotes inflammation. Finally, the implication of proposed model is discussed.
Collapse
Affiliation(s)
- Yu Cai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yangqiqi Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Shuo Leng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, China
| | - Yuanyuan Ma
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, 2799 W Grand Blvd, Detroit, MI 48202, USA
| | - Qiuting Wen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W.16th Street, Indianapolis, IN 46202-5188, USA
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Jiani Hu
- Department of Radiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
6
|
Djurich S, Secomb TW. Analysis of potassium ion diffusion from neurons to capillaries: Effects of astrocyte endfeet geometry. Eur J Neurosci 2024; 59:323-332. [PMID: 38123136 PMCID: PMC10872621 DOI: 10.1111/ejn.16232] [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: 06/06/2023] [Revised: 11/25/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Neurovascular coupling (NVC) refers to a local increase in cerebral blood flow in response to increased neuronal activity. Mechanisms of communication between neurons and blood vessels remain unclear. Astrocyte endfeet almost completely cover cerebral capillaries, suggesting that astrocytes play a role in NVC by releasing vasoactive substances near capillaries. An alternative hypothesis is that direct diffusion through the extracellular space of potassium ions (K+ ) released by neurons contributes to NVC. Here, the goal is to determine whether astrocyte endfeet present a barrier to K+ diffusion from neurons to capillaries. Two simplified 2D geometries of extracellular space, clefts between endfeet, and perivascular space are used: (i) a source 1 μm from a capillary; (ii) a neuron 15 μm from a capillary. K+ release is modelled as a step increase in [K+ ] at the outer boundary of the extracellular space. The time-dependent diffusion equation is solved numerically. In the first geometry, perivascular [K+ ] approaches its final value within 0.05 s. Decreasing endfeet cleft width or increasing perivascular space width slows the rise in [K+ ]. In the second geometry, the increase in perivascular [K+ ] occurs within 0.5 s and is insensitive to changes in cleft width or perivascular space width. Predicted levels of perivascular [K+ ] are sufficient to cause vasodilation, and the rise time is within the time for flow increase in NVC. These results suggest that direct diffusion of K+ through the extracellular space is a possible NVC signalling mechanism.
Collapse
Affiliation(s)
- Sara Djurich
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| | - Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
7
|
Bhattarai P, Gunasekaran TI, Reyes-Dumeyer D, Jülich D, Tayran H, Yilmaz E, Flaherty D, Lantigua R, Medrano M, Rivera D, Recio P, Ertekin-Taner N, Teich AF, Dickson DW, Holley S, Mayeux R, Kizil C, Vardarajan BN. Rare genetic variation in Fibronectin 1 ( FN1 ) protects against APOEe4 in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.02.573895. [PMID: 38260431 PMCID: PMC10802344 DOI: 10.1101/2024.01.02.573895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The risk of developing Alzheimer's disease (AD) significantly increases in individuals carrying the APOEε4 allele. Elderly cognitively healthy individuals with APOEε4 also exist, suggesting the presence of cellular mechanisms that counteract the pathological effects of APOEε4 ; however, these mechanisms are unknown. We hypothesized that APOEε4 carriers without dementia might carry genetic variations that could protect them from developing APOEε4- mediated AD pathology. To test this, we leveraged whole genome sequencing (WGS) data in National Institute on Aging Alzheimer's Disease Family Based Study (NIA-AD FBS), Washington Heights/Inwood Columbia Aging Project (WHICAP), and Estudio Familiar de Influencia Genetica en Alzheimer (EFIGA) cohorts and identified potentially protective variants segregating exclusively among unaffected APOEε4 carriers. In homozygous unaffected carriers above 70 years old, we identified 510 rare coding variants. Pathway analysis of the genes harboring these variants showed significant enrichment in extracellular matrix (ECM)-related processes, suggesting protective effects of functional modifications in ECM proteins. We prioritized two genes that were highly represented in the ECM-related gene ontology terms, (FN1) and collagen type VI alpha 2 chain ( COL6A2 ) and are known to be expressed at the blood-brain barrier (BBB), for postmortem validation and in vivo functional studies. The FN1 and COL6A2 protein levels were increased at the BBB in APOEε4 carriers with AD. Brain expression of cognitively unaffected homozygous APOEε4 carriers had significantly lower FN1 deposition and less reactive gliosis compared to homozygous APOEε4 carriers with AD, suggesting that FN1 might be a downstream driver of APOEε4 -mediated AD-related pathology and cognitive decline. To validate our findings, we used zebrafish models with loss-of-function (LOF) mutations in fn1b - the ortholog for human FN1 . We found that fibronectin LOF reduced gliosis, enhanced gliovascular remodeling and potentiated the microglial response, suggesting that pathological accumulation of FN1 could impair toxic protein clearance, which is ameliorated with FN1 LOF. Our study suggests vascular deposition of FN1 is related to the pathogenicity of APOEε4 , LOF variants in FN1 may reduce APOEε4 -related AD risk, providing novel clues to potential therapeutic interventions targeting the ECM to mitigate AD risk.
Collapse
|
8
|
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
|
9
|
Khaing ZZ, Chandrasekaran A, Katta A, Reed MJ. The Brain and Spinal Microvasculature in Normal Aging. J Gerontol A Biol Sci Med Sci 2023; 78:1309-1319. [PMID: 37093786 PMCID: PMC10395569 DOI: 10.1093/gerona/glad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Indexed: 04/25/2023] Open
Abstract
Changes in the brain and spinal cord microvasculature during normal aging contribute to the "sensitive" nature of aged central nervous system tissue to ischemic insults. In this review, we will examine alterations in the central nervous system microvasculature during normal aging, which we define as aging without a dominant pathology such as neurodegenerative processes, vascular injury or disease, or trauma. We will also discuss newer technologies to improve the study of central nervous system microvascular structure and function. Microvasculature within the brain and spinal cord will be discussed separately as anatomy and physiology differ between these compartments. Lastly, we will identify critical areas for future studies as well as key unanswered questions.
Collapse
Affiliation(s)
- Zin Z Khaing
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | | | - Anjali Katta
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | - May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Lee J, Thomas Broome S, Jansen MI, Mandwie M, Logan GJ, Marzagalli R, Musumeci G, Castorina A. Altered Hippocampal and Striatal Expression of Endothelial Markers and VIP/PACAP Neuropeptides in a Mouse Model of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:11118. [PMID: 37446298 DOI: 10.3390/ijms241311118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is one of the most common and severe manifestations of lupus; however, its pathogenesis is still poorly understood. While there is sparse evidence suggesting that the ongoing autoimmunity may trigger pathogenic changes to the central nervous system (CNS) microvasculature, culminating in inflammatory/ischemic damage, further evidence is still needed. In this study, we used the spontaneous mouse model of SLE (NZBWF1 mice) to investigate the expression of genes and proteins associated with endothelial (dys)function: tissue and urokinase plasminogen activators (tPA and uPA), intercellular and vascular adhesion molecules 1 (ICAM-1 and VCAM-1), brain derived neurotrophic factor (BDNF), endothelial nitric oxide synthase (eNOS) and Krüppel-like factor 4 (KLF4) and neuroprotection/immune modulation: pituitary adenylate cyclase-activating peptide (PACAP), vasoactive intestinal peptide (VIP), PACAP receptor (PAC1), VIP receptors 1 and 2 (VPAC1 and VPAC2). Analyses were carried out both in the hippocampus and striatum of SLE mice of two different age groups (2 and 7 months old), since age correlates with disease severity. In the hippocampus, we identified a gene/protein expression profile indicative of mild endothelial dysfunction, which increased in severity in aged SLE mice. These alterations were paralleled by moderate alterations in the expression of VIP, PACAP and related receptors. In contrast, we report a robust upregulation of endothelial activation markers in the striatum of both young and aged mice, concurrent with significant induction of the VIP/PACAP system. These data identify molecular signatures of endothelial alterations in the hippocampus and striatum of NZBWF1 mice, which are accompanied by a heightened expression of endogenous protective/immune-modulatory neuropeptides. Collectively, our results support the idea that NPSLE may cause alterations of the CNS micro-vascular compartment that cannot be effectively counteracted by the endogenous activity of the neuropeptides PACAP and VIP.
Collapse
Affiliation(s)
- Jayden Lee
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Sarah Thomas Broome
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Margo Iris Jansen
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Mawj Mandwie
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Grant J Logan
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Rubina Marzagalli
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95124 Catania, Italy
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
11
|
Kelly L, Sharp MM, Thomas I, Brown C, Schrag M, Antunes LV, Solopova E, Martinez-Gonzalez J, Rodríguez C, Carare RO. Targeting lysyl-oxidase (LOX) may facilitate intramural periarterial drainage for the treatment of Alzheimer's disease. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100171. [PMID: 37457664 PMCID: PMC10338210 DOI: 10.1016/j.cccb.2023.100171] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease is the commonest form of dementia. It is likely that a lack of clearance of amyloid beta (Aβ) results in its accumulation in the parenchyma as Aβ oligomers and insoluble plaques, and within the walls of blood vessels as cerebral amyloid angiopathy (CAA). The drainage of Aβ along the basement membranes of blood vessels as intramural periarterial drainage (IPAD), could be improved if the driving force behind IPAD could be augmented, therefore reducing Aβ accumulation. There are alterations in the composition of the vascular basement membrane in Alzheimer's disease. Lysyl oxidase (LOX) is an enzyme involved in the remodelling of the extracellular matrix and its expression and function is altered in various disease states. The expression of LOX is increased in Alzheimer's disease, but it is unclear whether this is a contributory factor in the impairment of IPAD in Alzheimer's disease. The pharmacological inhibition of LOX may be a strategy to improve IPAD and reduce the accumulation of Aβ in the parenchyma and within the walls of blood vessels.
Collapse
Affiliation(s)
- Louise Kelly
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | | | | | - Christopher Brown
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom, UK
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Lissa Ventura Antunes
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Elena Solopova
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - José Martinez-Gonzalez
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | | |
Collapse
|
12
|
Qi Z, Zhang C, Jian H, Hou M, Lou Y, Kang Y, Wang W, Lv Y, Shang S, Wang C, Li X, Feng S, Zhou H. N 1-Methyladenosine modification of mRNA regulates neuronal gene expression and oxygen glucose deprivation/reoxygenation induction. Cell Death Discov 2023; 9:159. [PMID: 37173310 PMCID: PMC10182019 DOI: 10.1038/s41420-023-01458-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
N1-Methyladenosine (m1A) is an abundant modification of transcripts, plays important roles in regulating mRNA structure and translation efficiency, and is dynamically regulated under stress. However, the characteristics and functions of mRNA m1A modification in primary neurons and oxygen glucose deprivation/reoxygenation (OGD/R) induced remain unclear. We first constructed a mouse cortical neuron OGD/R model and then used methylated RNA immunoprecipitation (MeRIP) and sequencing technology to demonstrate that m1A modification is abundant in neuron mRNAs and dynamically regulated during OGD/R induction. Our study suggests that Trmt10c, Alkbh3, and Ythdf3 may be m1A-regulating enzymes in neurons during OGD/R induction. The level and pattern of m1A modification change significantly during OGD/R induction, and differential methylation is closely associated with the nervous system. Our findings show that m1A peaks in cortical neurons aggregate at both the 5' and 3' untranslated regions. m1A modification can regulate gene expression, and peaks in different regions have different effects on gene expression. By analysing m1A-seq and RNA-seq data, we show a positive correlation between differentially methylated m1A peaks and gene expression. The correlation was verified by using qRT-PCR and MeRIP-RT-PCR. Moreover, we selected human tissue samples from Parkinson's disease (PD) and Alzheimer's disease (AD) patients from the Gene Expression Comprehensive (GEO) database to analyse the selected differentially expressed genes (DEGs) and differential methylation modification regulatory enzymes, respectively, and found similar differential expression results. We highlight the potential relationship between m1A modification and neuronal apoptosis following OGD/R induction. Furthermore, by mapping mouse cortical neurons and OGD/R-induced modification characteristics, we reveal the important role of m1A modification in OGD/R and gene expression regulation, providing new ideas for research on neurological damage.
Collapse
Affiliation(s)
- Zhangyang Qi
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Chi Zhang
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Huan Jian
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Mengfan Hou
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Yongfu Lou
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Yi Kang
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Wei Wang
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Yigang Lv
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Shenghui Shang
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China
| | - Chaoyu Wang
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China
| | - Xueying Li
- Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China.
| | - Hengxing Zhou
- Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, 300052, P.R. China.
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
13
|
Du F, Shusta EV, Palecek SP. Extracellular matrix proteins in construction and function of in vitro blood-brain barrier models. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2023.1130127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a highly impermeable barrier separating circulating blood and brain tissue. A functional BBB is critical for brain health, and BBB dysfunction has been linked to the pathophysiology of diseases such as stroke and Alzheimer’s disease. A variety of models have been developed to study the formation and maintenance of the BBB, ranging from in vivo animal models to in vitro models consisting of primary cells or cells differentiated from human pluripotent stem cells (hPSCs). These models must consider the composition and source of the cellular components of the neurovascular unit (NVU), including brain microvascular endothelial cells (BMECs), brain pericytes, astrocytes, and neurons, and how these cell types interact. In addition, the non-cellular components of the BBB microenvironment, such as the brain vascular basement membrane (BM) that is in direct contact with the NVU, also play key roles in BBB function. Here, we review how extracellular matrix (ECM) proteins in the brain vascular BM affect the BBB, with a particular focus on studies using hPSC-derived in vitro BBB models, and discuss how future studies are needed to advance our understanding of how the ECM affects BBB models to improve model performance and expand our knowledge on the formation and maintenance of the BBB.
Collapse
|
14
|
Evans TE, Knol MJ, Schwingenschuh P, Wittfeld K, Hilal S, Ikram MA, Dubost F, van Wijnen KMH, Katschnig P, Yilmaz P, de Bruijne M, Habes M, Chen C, Langer S, Völzke H, Ikram MK, Grabe HJ, Schmidt R, Adams HHH, Vernooij MW. Determinants of Perivascular Spaces in the General Population: A Pooled Cohort Analysis of Individual Participant Data. Neurology 2023; 100:e107-e122. [PMID: 36253103 PMCID: PMC9841448 DOI: 10.1212/wnl.0000000000201349] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Perivascular spaces (PVS) are emerging markers of cerebral small vessel disease (CSVD), but research on their determinants has been hampered by conflicting results from small single studies using heterogeneous rating methods. In this study, we therefore aimed to identify determinants of PVS burden in a pooled analysis of multiple cohort studies using 1 harmonized PVS rating method. METHODS Individuals from 10 population-based cohort studies with adult participants from the Uniform Neuro-Imaging of Virchow-Robin Spaces Enlargement consortium and the UK Biobank were included. On MRI scans, we counted PVS in 4 brain regions (mesencephalon, hippocampus, basal ganglia, and centrum semiovale) according to a uniform and validated rating protocol, both manually and automated using a deep learning algorithm. As potential determinants, we considered demographics, cardiovascular risk factors, APOE genotypes, and other imaging markers of CSVD. Negative binomial regression models were used to examine the association between these determinants and PVS counts. RESULTS In total, 39,976 individuals were included (age range 20-96 years). The average count of PVS in the 4 regions increased from the age 20 years (0-1 PVS) to 90 years (2-7 PVS). Men had more mesencephalic PVS (OR [95% CI] = 1.13 [1.08-1.18] compared with women), but less hippocampal PVS (0.82 [0.81-0.83]). Higher blood pressure, particularly diastolic pressure, was associated with more PVS in all regions (ORs between 1.04-1.05). Hippocampal PVS showed higher counts with higher high-density lipoprotein cholesterol levels (1.02 [1.01-1.02]), glucose levels (1.02 [1.01-1.03]), and APOE ε4-alleles (1.02 [1.01-1.04]). Furthermore, white matter hyperintensity volume and presence of lacunes were associated with PVS in multiple regions, but most strongly with the basal ganglia (1.13 [1.12-1.14] and 1.10 [1.09-1.12], respectively). DISCUSSION Various factors are associated with the burden of PVS, in part regionally specific, which points toward a multifactorial origin beyond what can be expected from PVS-related risk factor profiles. This study highlights the power of collaborative efforts in population neuroimaging research.
Collapse
Affiliation(s)
- Tavia E Evans
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Maria J Knol
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Petra Schwingenschuh
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Katharina Wittfeld
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Saima Hilal
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - M Arfan Ikram
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Florian Dubost
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Kimberlin M H van Wijnen
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Petra Katschnig
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Pinar Yilmaz
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Marleen de Bruijne
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Mohamad Habes
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Christopher Chen
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Sönke Langer
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Henry Völzke
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - M Kamran Ikram
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Hans J Grabe
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Reinhold Schmidt
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| | - Hieab H H Adams
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile.
| | - Meike W Vernooij
- From the Departments of Clinical Genetics (T.E.E., M.J.K., H.H.H.A.), Radiology and Nuclear Medicine (T.E.E., F.D., K.M.H.W., P.Y., M.B., H.H.H.A., M.W.V.), Epidemiology (M.J.K., M.A.I., P.Y., M.K.I., M.W.V.), and Neurology (M.K.I.), Erasmus MC, Rotterdam, the Netherlands; Department of Neurology (P.S., P.K., R.S.), Medical University of Graz, Austria; German Center for Neurodegenerative Diseases (DZNE) (K.W., M.H., H.J.G.), Site Rostock/Greifswald; Department of Psychiatry and Psychotherapy (K.W., H.J.G.) and Institute of Diagnostic Radiology and Neuroradiology (S.L.), University Medicine Greifswald, Germany; Department of Pharmacology (S.H., C.C.), National University of Singapore; Memory Aging & Cognition Centre (MACC) (S.H., C.C., M.K.I.), National University Health System, Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore; Department of Biomedical Data Sciences (F.D.), Stanford University, CA; J. Philip Kistler Stroke Research Center (P.Y.), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston; The Machine Learning Section (M.B.), Department of Computer Science, University of Copenhagen, Denmark; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC) (M.H.), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio (UTHSCSA), TX; and Latin American Brain Health (BrainLat) (H.H.H.A.), Universidad Adolfo Ibáñez, Santiago, Chile
| |
Collapse
|
15
|
Guo X, Zhang G, Peng Q, Huang L, Zhang Z, Zhang Z. Emerging Roles of Meningeal Lymphatic Vessels in Alzheimer's Disease. J Alzheimers Dis 2023; 94:S355-S366. [PMID: 36683509 PMCID: PMC10473149 DOI: 10.3233/jad-221016] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2022] [Indexed: 01/22/2023]
Abstract
Meningeal lymphatic vessels (mLVs), the functional lymphatic system present in the meninges, are the key drainage route responsible for the clearance of molecules, immune cells, and cellular debris from the cerebrospinal fluid and interstitial fluid into deep cervical lymph nodes. Aging and ApoE4, the two most important risk factors for Alzheimer's disease (AD), induce mLV dysfunction, decrease cerebrospinal fluid influx and outflux, and exacerbate amyloid pathology and cognitive dysfunction. Dysfunction of mLVs results in the deposition of metabolic products, accelerates neuroinflammation, and promotes the release of pro-inflammatory cytokines in the brain. Thus, mLVs represent a novel therapeutic target for treating neurodegenerative and neuroinflammatory diseases. This review aims to summarize the structure and function of mLVs and to discuss the potential effect of aging and ApoE4 on mLV dysfunction, as well as their roles in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xiaodi Guo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinyu Peng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liqin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
16
|
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: 3.5] [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
|
17
|
Anwar MM, Özkan E, Gürsoy-Özdemir Y. The role of extracellular matrix alterations in mediating astrocyte damage and pericyte dysfunction in Alzheimer's disease: A comprehensive review. Eur J Neurosci 2022; 56:5453-5475. [PMID: 34182602 DOI: 10.1111/ejn.15372] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
The brain is a highly vascularized tissue protected by the blood-brain barrier (BBB), a complex structure allowing only necessary substances to pass through into the brain while limiting the entrance of harmful toxins. The BBB comprises several components, and the most prominent features are tight junctions between endothelial cells (ECs), which are further wrapped in a layer of pericytes. Pericytes are multitasked cells embedded in a thick basement membrane (BM) that consists of a fibrous extracellular matrix (ECM) and are surrounded by astrocytic endfeet. The primary function of astrocytes and pericytes is to provide essential blood supply and vital nutrients to the brain. In Alzheimer's disease (AD), long-term neuroinflammatory cascades associated with infiltration of harmful neurotoxic proteins may lead to BBB dysfunction and altered ECM components resulting in brain homeostatic imbalance, synaptic damage, and declined cognitive functions. Moreover, BBB structure and functional integrity may be lost due to induced ECM alterations, astrocyte damage, and pericytes dysfunction, leading to amyloid-beta (Aβ) hallmarks deposition in different brain regions. Herein, we highlight how BBB, ECM, astrocytes, and pericytes dysfunction can play a leading role in AD's pathogenesis and discuss their impact on brain functions.
Collapse
Affiliation(s)
- Mai M Anwar
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority, Cairo, Egypt
| | - Esra Özkan
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Neuroscience Research Lab, Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.,Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
18
|
Sturmey E, Malaspina A. Blood biomarkers in ALS: challenges, applications and novel frontiers. Acta Neurol Scand 2022; 146:375-388. [PMID: 36156207 PMCID: PMC9828487 DOI: 10.1111/ane.13698] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/12/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease among adults. With diagnosis reached relatively late into the disease process, extensive motor cell loss narrows the window for therapeutic opportunities. Clinical heterogeneity in ALS and the lack of disease-specific biomarkers have so far led to large-sized clinical trials with long follow-up needed to define clinical outcomes. In advanced ALS patients, there is presently limited scope to use imaging or invasive cerebrospinal fluid (CSF) collection as a source of disease biomarkers. The development of more patient-friendly and accessible blood biomarker assays is hampered by analytical hurdles like the matrix effect of blood components. However, blood also provides the opportunity to identify disease-specific adaptive changes of the stoichiometry and conformation of target proteins and the endogenous immunological response to low-abundance brain peptides, such as neurofilaments (Nf). Among those biomarkers under investigation in ALS, the change in concentration before or after diagnosis of Nf has been shown to aid prognostication and to allow the a priori stratification of ALS patients into smaller sized and clinically more homogeneous cohorts, supporting more affordable clinical trials. Here, we discuss the technical hurdles affecting reproducible and sensitive biomarker measurement in blood. We also summarize the state of the art of non-CSF biomarkers in the study of prognosis, disease progression, and treatment response. We will then address the potential as disease-specific biomarkers of the newly discovered cryptic peptides which are formed down-stream of TDP-43 loss of function, the hallmark of ALS pathobiology.
Collapse
Affiliation(s)
- Ellie Sturmey
- Centre of Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK
| | - Andrea Malaspina
- Centre of Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK.,Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
19
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
20
|
Xiong W, Cai J, Li R, Wen C, Tan H. Rare Variant Analysis and Molecular Dynamics Simulation in Alzheimer’s Disease Identifies Exonic Variants in FLG. Genes (Basel) 2022; 13. [PMID: 35627223 PMCID: PMC9141140 DOI: 10.3390/genes13050838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Although an increasing number of common variants contributing to Alzheimer’s disease (AD) are uncovered by genome-wide association studies, they can only explain less than half of the heritability of AD. Rare variant association studies (RVAS) has become an increasingly important area to explain the risk or trait variability of AD. Method: To investigate the potential rare variants that cause AD, we screened 70,209 rare variants from two cohorts of a 175 AD cohort and a 214 cognitively normal cohort from the Alzheimer’s Disease Neuroimaging Initiative database. MIRARE, a novel RVAS method, was performed on 232 non-synonymous variants selected by ANNOVAR annotation. Molecular docking and molecular dynamics (MD) simulation were adopted to verify the interaction between the chosen functional variants and BACE1. Results: MIRAGE analysis revealed significant associations between AD and six potential pathogenic genes, including PREX2, FLG, DHX16, NID2, ZnF585B and ZnF875. Only interactions between FLG (including wild type and rs3120654(SER742TYR)) and BACE1 were verified by molecular docking and MD simulation. The interaction of FLG(SER742TYR) with BACE1 was greater than that of wildtype FLG with BACE1. Conclusions: According to the literature search, bio-informatics analysis, and molecular docking and MD simulation, we find non-synonymous rare variants in six genes, especially FLG(rs3120654), that may play key roles in AD.
Collapse
Affiliation(s)
- Weixue Xiong
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | - Jiahui Cai
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | - Ruijia Li
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei 230000, China;
| | - Canhong Wen
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei 230000, China
| | - Haizhu Tan
- Department of Preventive Medicine, Shantou University Medical College, Shantou 515000, China
| | | |
Collapse
|
21
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
22
|
Nguyen B, Bix G, Yao Y. Basal lamina changes in neurodegenerative disorders. Mol Neurodegener 2021; 16:81. [PMID: 34876200 PMCID: PMC8650282 DOI: 10.1186/s13024-021-00502-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neurodegenerative disorders are a group of age-associated diseases characterized by progressive degeneration of the structure and function of the CNS. Two key pathological features of these disorders are blood-brain barrier (BBB) breakdown and protein aggregation. MAIN BODY The BBB is composed of various cell types and a non-cellular component---the basal lamina (BL). Although how different cells affect the BBB is well studied, the roles of the BL in BBB maintenance and function remain largely unknown. In addition, located in the perivascular space, the BL is also speculated to regulate protein clearance via the meningeal lymphatic/glymphatic system. Recent studies from our laboratory and others have shown that the BL actively regulates BBB integrity and meningeal lymphatic/glymphatic function in both physiological and pathological conditions, suggesting that it may play an important role in the pathogenesis and/or progression of neurodegenerative disorders. In this review, we focus on changes of the BL and its major components during aging and in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). First, we introduce the vascular and lymphatic systems in the CNS. Next, we discuss the BL and its major components under homeostatic conditions, and summarize their changes during aging and in AD, PD, and ALS in both rodents and humans. The functional significance of these alterations and potential therapeutic targets are also reviewed. Finally, key challenges in the field and future directions are discussed. CONCLUSIONS Understanding BL changes and the functional significance of these changes in neurodegenerative disorders will fill the gap of knowledge in the field. Our goal is to provide a clear and concise review of the complex relationship between the BL and neurodegenerative disorders to stimulate new hypotheses and further research in this field.
Collapse
Affiliation(s)
- Benjamin Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Gregory Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA.
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, Tampa, Florida, 33612, USA.
| |
Collapse
|
23
|
Gireud-Goss M, Mack AF, McCullough LD, Urayama A. Cerebral Amyloid Angiopathy and Blood-Brain Barrier Dysfunction. Neuroscientist 2021; 27:668-684. [PMID: 33238806 PMCID: PMC9853919 DOI: 10.1177/1073858420954811] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cerebral hemorrhage, a devastating subtype of stroke, is often caused by hypertension and cerebral amyloid angiopathy (CAA). Pathological evidence of CAA is detected in approximately half of all individuals over the age of 70 and is associated with cortical microinfarcts and cognitive impairment. The underlying pathophysiology of CAA is characterized by accumulation of pathogenic amyloid β (Aβ) fragments of amyloid precursor protein in the cerebral vasculature. Vascular deposition of Aβ damages the vessel wall, results in blood-brain barrier (BBB) leakiness, vessel occlusion or rupture, and leads to hemorrhages and decreased cerebral blood flow that negatively affects vessel integrity and cognitive function. Currently, the main hypothesis surrounding the mechanism of CAA pathogenesis is that there is an impaired clearance of Aβ peptides, which includes compromised perivascular drainage as well as dysfunction of BBB transport. Also, the immune response in CAA pathogenesis plays an important role. Therefore, the mechanism by which Aβ vascular deposition occurs is crucial for our understanding of CAA pathogenesis and for the development of potential therapeutic options.
Collapse
Affiliation(s)
- Monica Gireud-Goss
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexis F. Mack
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
24
|
The α-dystrobrevins play a key role in maintaining the structure and function of the extracellular matrix-significance for protein elimination failure arteriopathies. Acta Neuropathol Commun 2021; 9:171. [PMID: 34674769 PMCID: PMC8532274 DOI: 10.1186/s40478-021-01274-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
The extracellular matrix (ECM) of the cerebral vasculature provides a pathway for the flow of interstitial fluid (ISF) and solutes out of the brain by intramural periarterial drainage (IPAD). Failure of IPAD leads to protein elimination failure arteriopathies such as cerebral amyloid angiopathy (CAA). The ECM consists of a complex network of glycoproteins and proteoglycans that form distinct basement membranes (BM) around different vascular cell types. Astrocyte endfeet that are localised against the walls of blood vessels are tethered to these BMs by dystrophin associated protein complex (DPC). Alpha-dystrobrevin (α-DB) is a key dystrophin associated protein within perivascular astrocyte endfeet; its deficiency leads to a reduction in other dystrophin associated proteins, loss of AQP4 and altered ECM. In human dementia cohorts there is a positive correlation between dystrobrevin gene expression and CAA. In the present study, we test the hypotheses that (a) the positive correlation between dystrobrevin gene expression and CAA is associated with elevated expression of α-DB at glial-vascular endfeet and (b) a deficiency in α-DB results in changes to the ECM and failure of IPAD. We used human post-mortem brain tissue with different severities of CAA and transgenic α-DB deficient mice. In human post-mortem tissue we observed a significant increase in vascular α-DB with CAA (CAA vrs. Old p < 0.005, CAA vrs. Young p < 0.005). In the mouse model of α-DB deficiency, there was early modifications to vascular ECM (collagen IV and BM thickening) that translated into reduced IPAD efficiency. Our findings highlight the important role of α-DB in maintaining structure and function of ECM, particularly as a pathway for the flow of ISF and solutes out of the brain by IPAD.
Collapse
|
25
|
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: 2.3] [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
|
26
|
Hussain B, Fang C, Chang J. Blood-Brain Barrier Breakdown: An Emerging Biomarker of Cognitive Impairment in Normal Aging and Dementia. Front Neurosci 2021; 15:688090. [PMID: 34489623 PMCID: PMC8418300 DOI: 10.3389/fnins.2021.688090] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The blood–brain barrier (BBB) plays a vital role in maintaining the specialized microenvironment of the neural tissue. It separates the peripheral circulatory system from the brain parenchyma while facilitating communication. Alterations in the distinct physiological properties of the BBB lead to BBB breakdown associated with normal aging and various neurodegenerative diseases. In this review, we first briefly discuss the aging process, then review the phenotypes and mechanisms of BBB breakdown associated with normal aging that further cause neurodegeneration and cognitive impairments. We also summarize dementia such as Alzheimer's disease (AD) and vascular dementia (VaD) and subsequently discuss the phenotypes and mechanisms of BBB disruption in dementia correlated with cognition decline. Overlaps between AD and VaD are also discussed. Techniques that could identify biomarkers associated with BBB breakdown are briefly summarized. Finally, we concluded that BBB breakdown could be used as an emerging biomarker to assist to diagnose cognitive impairment associated with normal aging and dementia.
Collapse
Affiliation(s)
- Basharat Hussain
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Fang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
27
|
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: 22] [Impact Index Per Article: 7.3] [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
|
28
|
Mutimer CA, Keragala CB, Markus HS, Werring DJ, Cloud GC, Medcalf RL. Cerebral Amyloid Angiopathy and the Fibrinolytic System: Is Plasmin a Therapeutic Target? Stroke 2021; 52:2707-2714. [PMID: 34126761 DOI: 10.1161/strokeaha.120.033107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral amyloid angiopathy is a devastating cause of intracerebral hemorrhage for which there is no specific secondary stroke prevention treatment. Here we review the current literature regarding cerebral amyloid angiopathy pathophysiology and treatment, as well as what is known of the fibrinolytic pathway and its interaction with amyloid. We postulate that tranexamic acid is a potential secondary stroke prevention treatment agent in sporadic cerebral amyloid angiopathy, although further research is required.
Collapse
Affiliation(s)
- Chloe A Mutimer
- Department of Neurology, Alfred Hospital, Melbourne, Australia (C.A.M., G.C.C.)
| | - Charithani B Keragala
- Australian Centre for Blood Diseases (C.B.K., R.L.M.), Monash University, Melbourne, Australia
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge, United Kingdom (H.S.M.)
| | - David J Werring
- Stroke Research Centre, Queen Square Institute of Neurology, London, United Kingdom (D.J.W.)
| | - Geoffrey C Cloud
- Department of Neurology, Alfred Hospital, Melbourne, Australia (C.A.M., G.C.C.).,Department of Clinical Neuroscience, Central Clinical School (G.C.C.), Monash University, Melbourne, Australia
| | - Robert L Medcalf
- Australian Centre for Blood Diseases (C.B.K., R.L.M.), Monash University, Melbourne, Australia
| |
Collapse
|
29
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
30
|
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: 2.0] [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
|
31
|
Abstract
The blood-brain barrier (BBB) protects the central nervous system (CNS) from unregulated exposure to the blood and its contents. The BBB also controls the blood-to-brain and brain-to-blood permeation of many substances, resulting in nourishment of the CNS, its homeostatic regulation and communication between the CNS and peripheral tissues. The cells forming the BBB communicate with cells of the brain and in the periphery. This highly regulated interface changes with healthy aging. Here, we review those changes, starting with morphology and disruption. Transporter changes include those for amyloid beta peptide, glucose and drugs. Brain fluid dynamics, pericyte health and basement membrane and glycocalyx compositions are all altered with healthy aging. Carrying the ApoE4 allele leads to an acceleration of most of the BBB's age-related changes. We discuss how alterations in the BBB that occur with healthy aging reflect adaptation to the postreproductive phase of life and may affect vulnerability to age-associated diseases.
Collapse
|
32
|
Nizari S, Wells JA, Carare RO, Romero IA, Hawkes CA. Loss of cholinergic innervation differentially affects eNOS-mediated blood flow, drainage of Aβ and cerebral amyloid angiopathy in the cortex and hippocampus of adult mice. Acta Neuropathol Commun 2021; 9:12. [PMID: 33413694 PMCID: PMC7791879 DOI: 10.1186/s40478-020-01108-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/15/2020] [Indexed: 11/18/2022] Open
Abstract
Vascular dysregulation and cholinergic basal forebrain degeneration are both early pathological events in the development of Alzheimer’s disease (AD). Acetylcholine contributes to localised arterial dilatation and increased cerebral blood flow (CBF) during neurovascular coupling via activation of endothelial nitric oxide synthase (eNOS). Decreased vascular reactivity is suggested to contribute to impaired clearance of β-amyloid (Aβ) along intramural periarterial drainage (IPAD) pathways of the brain, leading to the development of cerebral amyloid angiopathy (CAA). However, the possible relationship between loss of cholinergic innervation, impaired vasoreactivity and reduced clearance of Aβ from the brain has not been previously investigated. In the present study, intracerebroventricular administration of mu-saporin resulted in significant death of cholinergic neurons and fibres in the medial septum, cortex and hippocampus of C57BL/6 mice. Arterial spin labelling MRI revealed a loss of CBF response to stimulation of eNOS by the Rho-kinase inhibitor fasudil hydrochloride in the cortex of denervated mice. By contrast, the hippocampus remained responsive to drug treatment, in association with altered eNOS expression. Fasudil hydrochloride significantly increased IPAD in the hippocampus of both control and saporin-treated mice, while increased clearance from the cortex was only observed in control animals. Administration of mu-saporin in the TetOAPPSweInd mouse model of AD was associated with a significant and selective increase in Aβ40-positive CAA. These findings support the importance of the interrelationship between cholinergic innervation and vascular function in the aetiology and/or progression of CAA and suggest that combined eNOS/cholinergic therapies may improve the efficiency of Aβ removal from the brain and reduce its deposition as CAA.
Collapse
|
33
|
Young KZ, Xu G, Keep SG, Borjigin J, Wang MM. Overlapping Protein Accumulation Profiles of CADASIL and CAA: Is There a Common Mechanism Driving Cerebral Small-Vessel Disease? THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:1871-1887. [PMID: 33387456 DOI: 10.1016/j.ajpath.2020.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and cerebral amyloid angiopathy (CAA) are two distinct vascular angiopathies that share several similarities in clinical presentation and vascular pathology. Given the clinical and pathologic overlap, the molecular overlap between CADASIL and CAA was explored. CADASIL and CAA protein profiles from recently published proteomics-based and immuno-based studies were compared to investigate the potential for shared disease mechanisms. A comparison of affected proteins in each disease highlighted 19 proteins that are regulated in both CADASIL and CAA. Functional analysis of the shared proteins predicts significant interaction between them and suggests that most enriched proteins play roles in extracellular matrix structure and remodeling. Proposed models to explain the observed enrichment of extracellular matrix proteins include both increased protein secretion and decreased protein turnover by sequestration of chaperones and proteases or formation of stable protein complexes. Single-cell RNA sequencing of vascular cells in mice suggested that the vast majority of the genes accounting for the overlapped proteins between CADASIL and CAA are expressed by fibroblasts. Thus, our current understanding of the molecular profiles of CADASIL and CAA appears to support potential for common mechanisms underlying the two disorders.
Collapse
Affiliation(s)
- Kelly Z Young
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Gang Xu
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Simon G Keep
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan
| | - Jimo Borjigin
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Michael M Wang
- Departments of Neurology, University of Michigan, Ann Arbor, Michigan; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan.
| |
Collapse
|
34
|
Howe MD, McCullough LD, Urayama A. The Role of Basement Membranes in Cerebral Amyloid Angiopathy. Front Physiol 2020; 11:601320. [PMID: 33329053 PMCID: PMC7732667 DOI: 10.3389/fphys.2020.601320] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Dementia is a neuropsychiatric syndrome characterized by cognitive decline in multiple domains, often leading to functional impairment in activities of daily living, disability, and death. The most common causes of age-related progressive dementia include Alzheimer's disease (AD) and vascular cognitive impairment (VCI), however, mixed disease pathologies commonly occur, as epitomized by a type of small vessel pathology called cerebral amyloid angiopathy (CAA). In CAA patients, the small vessels of the brain become hardened and vulnerable to rupture, leading to impaired neurovascular coupling, multiple microhemorrhage, microinfarction, neurological emergencies, and cognitive decline across multiple functional domains. While the pathogenesis of CAA is not well understood, it has long been thought to be initiated in thickened basement membrane (BM) segments, which contain abnormal protein deposits and amyloid-β (Aβ). Recent advances in our understanding of CAA pathogenesis link BM remodeling to functional impairment of perivascular transport pathways that are key to removing Aβ from the brain. Dysregulation of this process may drive CAA pathogenesis and provides an important link between vascular risk factors and disease phenotype. The present review summarizes how the structure and composition of the BM allows for perivascular transport pathways to operate in the healthy brain, and then outlines multiple mechanisms by which specific dementia risk factors may promote dysfunction of perivascular transport pathways and increase Aβ deposition during CAA pathogenesis. A better understanding of how BM remodeling alters perivascular transport could lead to novel diagnostic and therapeutic strategies for CAA patients.
Collapse
Affiliation(s)
| | | | - Akihiko Urayama
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
35
|
Damodarasamy M, Vernon RB, Pathan JL, Keene CD, Day AJ, Banks WA, Reed MJ. The microvascular extracellular matrix in brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA). Fluids Barriers CNS 2020; 17:60. [PMID: 32993718 PMCID: PMC7525948 DOI: 10.1186/s12987-020-00219-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/09/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The microvasculature (MV) of brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA), in the absence of concurrent pathologies (e.g., infarctions, Lewy bodies), is incompletely understood. OBJECTIVE To analyze microvascular density, diameter and extracellular matrix (ECM) content in association with ADNC and CAA. METHODS We examined samples of cerebral cortex and isolated brain microvasculature (MV) from subjects with the National Institute on Aging-Alzheimer's Association (NIA-AA) designations of not-, intermediate-, or high ADNC and from subjects with no CAA and moderate-severe CAA. Cases for all groups were selected with no major (territorial) strokes, ≤ 1 microinfarct in screening sections, and no Lewy body pathology. MV density and diameter were measured from cortical brain sections. Levels of basement membrane (BM) ECM components, the protein product of TNF-stimulated gene-6 (TSG-6), and the ubiquitous glycosaminoglycan hyaluronan (HA) were assayed by western blots or HA ELISA of MV lysates. RESULTS We found no significant changes in MV density or diameter among any of the groups. Levels of BM laminin and collagen IV (col IV) were lower in MV isolated from the high ADNC vs. not-ADNC groups. In contrast, BM laminin was significantly higher in MV from the moderate-severe CAA vs. the no CAA groups. TSG-6 and HA content were higher in the presence of both high ADNC and CAA, whereas levels of BM fibronectin and perlecan were similar among all groups. CONCLUSIONS Cortical MV density and diameter are not appreciably altered by ADNC or CAA. TSG-6 and HA are increased in both ADNC and CAA, with laminin and col IV decreased in the BM of high ADNC, but laminin increased in moderate-severe CAA. These results show that changes in the ECM occur in AD and CAA, but independently of one another, and likely reflect on the regional functioning of the brain microvasculature.
Collapse
Affiliation(s)
- Mamatha Damodarasamy
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Robert B Vernon
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jasmine L Pathan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA, USA
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - May J Reed
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA.
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington Harborview Medical Center, Seattle, WA, 98104, USA.
| |
Collapse
|
36
|
Frost M, Keable A, Baseley D, Sealy A, Andreea Zbarcea D, Gatherer M, Yuen HM, Sharp MM, Weller RO, Attems J, Smith C, Chiarot PR, Carare RO. Vascular α1A Adrenergic Receptors as a Potential Therapeutic Target for IPAD in Alzheimer's Disease. Pharmaceuticals (Basel) 2020; 13:ph13090261. [PMID: 32971843 PMCID: PMC7560129 DOI: 10.3390/ph13090261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 01/20/2023] Open
Abstract
Drainage of interstitial fluid from the brain occurs via the intramural periarterial drainage (IPAD) pathways along the basement membranes of cerebral capillaries and arteries against the direction of blood flow into the brain. The cerebrovascular smooth muscle cells (SMCs) provide the motive force for driving IPAD, and their decrease in function may explain the deposition of amyloid-beta as cerebral amyloid angiopathy (CAA), a key feature of Alzheimer’s disease. The α-adrenoceptor subtype α1A is abundant in the brain, but its distribution in the cerebral vessels is unclear. We analysed cultured human cerebrovascular SMCs and young, old and CAA human brains for (a) the presence of α1A receptor and (b) the distribution of the α1A receptor within the cerebral vessels. The α1A receptor was present on the wall of cerebrovascular SMCs. No significant changes were observed in the vascular expression of the α1A-adrenergic receptor in young, old and CAA cases. The pattern of vascular staining appeared less punctate and more diffuse with ageing and CAA. Our results show that the α1A-adrenergic receptor is preserved in cerebral vessels with ageing and in CAA and is expressed on cerebrovascular smooth muscle cells, suggesting that vascular adrenergic receptors may hold potential for therapeutic targeting of IPAD.
Collapse
Affiliation(s)
- Miles Frost
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Abby Keable
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Dan Baseley
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Amber Sealy
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Diana Andreea Zbarcea
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Maureen Gatherer
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Ho Ming Yuen
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Matt MacGregor Sharp
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Roy O. Weller
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Newacstle upon Tyne NE4 5PL, UK;
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK;
| | - Paul R. Chiarot
- Department of Mechanical Engineering, State University of New York at Binghamton, Binghamton, NY 13902, USA;
| | - Roxana O. Carare
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (M.F.); (A.K.); (D.B.); (A.S.); (D.A.Z.); (M.G.); (H.M.Y.); (M.M.S.); (R.O.W.)
- Correspondence:
| |
Collapse
|
37
|
Demonstrating a reduced capacity for removal of fluid from cerebral white matter and hypoxia in areas of white matter hyperintensity associated with age and dementia. Acta Neuropathol Commun 2020; 8:131. [PMID: 32771063 PMCID: PMC7414710 DOI: 10.1186/s40478-020-01009-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
White matter hyperintensities (WMH) occur in association with dementia but the aetiology is unclear. Here we test the hypothesis that there is a combination of impaired elimination of interstitial fluid from the white matter together with a degree of hypoxia in WMH. One of the mechanisms for the elimination of amyloid-β (Aβ) from the brain is along the basement membranes in the walls of capillaries and arteries (Intramural Peri-Arterial Drainage – IPAD). We compared the dynamics of IPAD in the grey matter of the hippocampus and in the white matter of the corpus callosum in 10 week old C57/B16 mice by injecting soluble Aβ as a tracer. The dynamics of IPAD in the white matter were significantly slower compared with the grey matter and this was associated with a lower density of capillaries in the white matter. Exposing cultures of smooth muscle cells to hypercapnia as a model of cerebral hypoperfusion resulted in a reduction in fibronectin and an increase in laminin in the extracellular matrix. Similar changes were detected in the white matter in human WMH suggesting that hypercapnia/hypoxia may play a role in WMH. Employing therapies to enhance both IPAD and blood flow in the white matter may reduce WMH in patients with dementia.
Collapse
|
38
|
Carare RO, Aldea R, Agarwal N, Bacskai BJ, Bechman I, Boche D, Bu G, Bulters D, Clemens A, Counts SE, de Leon M, Eide PK, Fossati S, Greenberg SM, Hamel E, Hawkes CA, Koronyo‐Hamaoui M, Hainsworth AH, Holtzman D, Ihara M, Jefferson A, Kalaria RN, Kipps CM, Kanninen KM, Leinonen V, McLaurin J, Miners S, Malm T, Nicoll JAR, Piazza F, Paul G, Rich SM, Saito S, Shih A, Scholtzova H, Snyder H, Snyder P, Thormodsson FR, van Veluw SJ, Weller RO, Werring DJ, Wilcock D, Wilson MR, Zlokovic BV, Verma A. Clearance of interstitial fluid (ISF) and CSF (CLIC) group-part of Vascular Professional Interest Area (PIA): Cerebrovascular disease and the failure of elimination of Amyloid-β from the brain and retina with age and Alzheimer's disease-Opportunities for Therapy. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12053. [PMID: 32775596 PMCID: PMC7396859 DOI: 10.1002/dad2.12053] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
Two of the key functions of arteries in the brain are (1) the well-recognized supply of blood via the vascular lumen and (2) the emerging role for the arterial walls as routes for the elimination of interstitial fluid (ISF) and soluble metabolites, such as amyloid beta (Aβ), from the brain and retina. As the brain and retina possess no conventional lymphatic vessels, fluid drainage toward peripheral lymph nodes is mediated via transport along basement membranes in the walls of capillaries and arteries that form the intramural peri-arterial drainage (IPAD) system. IPAD tends to fail as arteries age but the mechanisms underlying the failure are unclear. In some people this is reflected in the accumulation of Aβ plaques in the brain in Alzheimer's disease (AD) and deposition of Aβ within artery walls as cerebral amyloid angiopathy (CAA). Knowledge of the dynamics of IPAD and why it fails with age is essential for establishing diagnostic tests for the early stages of the disease and for devising therapies that promote the clearance of Aβ in the prevention and treatment of AD and CAA. This editorial is intended to introduce the rationale that has led to the establishment of the Clearance of Interstitial Fluid (ISF) and CSF (CLIC) group, within the Vascular Professional Interest Area of the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Diederik Bulters
- University of SouthamptonSouthamptonUK
- University Hospital Southampton NHS TrustSouthamptonUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christopher M. Kipps
- University of SouthamptonSouthamptonUK
- University Hospital Southampton NHS TrustSouthamptonUK
| | | | | | | | | | - Tarja Malm
- University of Eastern FinlandKuopioFinland
| | | | | | | | | | - Satoshi Saito
- National Cerebral and Cardiovascular CenterOsakaJapan
| | - Andy Shih
- Seattle Children's HospitalSeattleWashingtonUSA
| | | | | | - Peter Snyder
- University of Rhode IslandSouth KingstownRhode IslandUSA
| | | | | | | | - David J. Werring
- Stroke Research CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | | | | | | | - Ajay Verma
- CODIAK BiosciencesCambridgeMassachusettsUSA
| |
Collapse
|
39
|
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: 3.5] [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
|
40
|
Ma J, Ma C, Li J, Sun Y, Ye F, Liu K, Zhang H. Extracellular Matrix Proteins Involved in Alzheimer's Disease. Chemistry 2020; 26:12101-12110. [PMID: 32207199 DOI: 10.1002/chem.202000782] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/22/2020] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases and characterized by cognitive and memory impairments. Emerging evidence suggests that the extracellular matrix (ECM) in the brain plays an important role in the etiology of AD. It has been detected that the levels of ECM proteins have changed in the brains of AD patients and animal models. Some ECM components, for example, elastin and heparan sulfate proteoglycans, are considered to promote the upregulation of extracellular amyloid-beta (Aβ) proteins. In addition, collagen VI and laminin are shown to have interactions with Aβ peptides, which might lead to the clearance of those peptides. Thus, ECM proteins are involved in both amyloidosis and neuroprotection in the AD process. However, the molecular mechanism of neuronal ECM proteins on the pathophysiology of AD remains elusive. More investigation of ECM proteins with AD pathogenesis is needed, and this may lead to novel therapeutic strategies and biomarkers for AD.
Collapse
Affiliation(s)
- Jun Ma
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, P.R. China.,State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Chao Ma
- School of Engineering and Applied Sciences & Department of Physics, Harvard University, 29 Oxford Street, Cambridge, MA, 02138, USA
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Yao Sun
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China
| | - Fangfu Ye
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, P.R. China.,Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China.,Department of Chemistry, Tsinghua University, Beijing, 100084, P.R. China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P.R. China.,Department of Chemistry, Tsinghua University, Beijing, 100084, P.R. China
| |
Collapse
|
41
|
Kumar V, Lee JD, Coulson EJ, Woodruff TM. A validated quantitative method for the assessment of neuroprotective barrier impairment in neurodegenerative disease models. J Neurochem 2020; 158:807-817. [PMID: 32628780 DOI: 10.1111/jnc.15119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB) are highly specialized structures that limit molecule entry from the blood and maintain homeostasis within the central nervous system (CNS). BBB and BSCB breakdown are associated with multiple neurodegenerative diseases. Given the key role of neuroprotective barrier impairment in neurodegeneration, it is important to identify an effective quantitative method to assess barrier integrity in animal models. In this study, we developed and validated a quantitative method for assessing BBB and BSCB integrity using sodium fluorescein, a compound that outperformed other fluorescent dyes. We demonstrated using this method that multiple CNS regions progressively increase in permeability in models of Huntington's disease and amyotrophic lateral sclerosis, whereas biphasic disruption occurred in a mouse model of Alzheimer's disease with disease progression. Collectively, we report a quantitative fluorometric marker with validated reproducible experimental methods that allows the effective assessment of BBB and BSCB integrity in animal models. This method could be useful to further the understanding of the contribution of these neuroprotective barriers to neurodegeneration processes.
Collapse
Affiliation(s)
- Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - Elizabeth J Coulson
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Qld, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Qld, Australia
| |
Collapse
|
42
|
Ingala S, Mazzai L, Sudre CH, Salvadó G, Brugulat-Serrat A, Wottschel V, Falcon C, Operto G, Tijms B, Gispert JD, Molinuevo JL, Barkhof F. The relation between APOE genotype and cerebral microbleeds in cognitively unimpaired middle- and old-aged individuals. Neurobiol Aging 2020; 95:104-114. [PMID: 32791423 DOI: 10.1016/j.neurobiolaging.2020.06.015] [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: 11/05/2019] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 01/10/2023]
Abstract
Positive associations between cerebral microbleeds (CMBs) and APOE-ε4 (apolipoprotein E) genotype have been reported in Alzheimer's disease, but show conflicting results. We investigated the effect of APOE genotype on CMBs in a cohort of cognitively unimpaired middle- and old-aged individuals enriched for APOE-ε4 genotype. Participants from ALFA (Alzheimer and Families) cohort were included and their magnetic resonance scans assessed (n = 564, 50% APOE-ε4 carriers). Quantitative magnetic resonance analyses included visual ratings, atrophy measures, and white matter hyperintensity (WMH) segmentations. The prevalence of CMBs was 17%, increased with age (p < 0.05), and followed an increasing trend paralleling APOE-ε4 dose. The number of CMBs was significantly higher in APOE-ε4 homozygotes compared to heterozygotes and non-carriers (p < 0.05). This association was driven by lobar CMBs (p < 0.05). CMBs co-localized with WMH (p < 0.05). No associations between CMBs and APOE-ε2, gray matter volumes, and cognitive performance were found. Our results suggest that cerebral vessels of APOE-ε4 homozygous are more fragile, especially in lobar locations. Co-occurrence of CMBs and WMH suggests that such changes localize in areas with increased vascular vulnerability.
Collapse
Affiliation(s)
- Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - Linda Mazzai
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Department of Medicine (DiMED), Institute of Radiology, University of Padua, Padua, Italy
| | - Carole H Sudre
- Engineering and Imaging Sciences, King's College London, London, UK; Dementia Research Centre, University College London, London, UK; Centre for Medical Imaging Computing, Faculty of Engineering, University College London, London, UK
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
| | - Viktor Wottschel
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain
| | - Betty Tijms
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain; Pompeu Fabra University, Barcelona, Spain.
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Pompeu Fabra University, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; Institutes of Neurology and Healthcare Engineering, UCL, London, UK
| | | |
Collapse
|
43
|
Troili F, Cipollini V, Moci M, Morena E, Palotai M, Rinaldi V, Romano C, Ristori G, Giubilei F, Salvetti M, Orzi F, Guttmann CRG, Cavallari M. Perivascular Unit: This Must Be the Place. The Anatomical Crossroad Between the Immune, Vascular and Nervous System. Front Neuroanat 2020; 14:17. [PMID: 32372921 PMCID: PMC7177187 DOI: 10.3389/fnana.2020.00017] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/23/2020] [Indexed: 12/25/2022] Open
Abstract
Most neurological disorders seemingly have heterogenous pathogenesis, with overlapping contribution of neuronal, immune and vascular mechanisms of brain injury. The perivascular space in the brain represents a crossroad where those mechanisms interact, as well as a key anatomical component of the recently discovered glymphatic pathway, which is considered to play a crucial role in the clearance of brain waste linked to neurodegenerative diseases. The pathological interplay between neuronal, immune and vascular factors can create an environment that promotes self-perpetration of mechanisms of brain injury across different neurological diseases, including those that are primarily thought of as neurodegenerative, neuroinflammatory or cerebrovascular. Changes of the perivascular space can be monitored in humans in vivo using magnetic resonance imaging (MRI). In the context of glymphatic clearance, MRI-visible enlarged perivascular spaces (EPVS) are considered to reflect glymphatic stasis secondary to the perivascular accumulation of brain debris, although they may also represent an adaptive mechanism of the glymphatic system to clear them. EPVS are also established correlates of dementia and cerebral small vessel disease (SVD) and are considered to reflect brain inflammatory activity. In this review, we describe the “perivascular unit” as a key anatomical and functional substrate for the interaction between neuronal, immune and vascular mechanisms of brain injury, which are shared across different neurological diseases. We will describe the main anatomical, physiological and pathological features of the perivascular unit, highlight potential substrates for the interplay between different noxae and summarize MRI studies of EPVS in cerebrovascular, neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Fernanda Troili
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Virginia Cipollini
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Marco Moci
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Neuroscience Section, University of Salerno, Baronissi, Italy
| | - Emanuele Morena
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Miklos Palotai
- Harvard Medical School, Center for Neurological Imaging, Brigham and Women's Hospital, Boston, MA, United States
| | - Virginia Rinaldi
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Carmela Romano
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Giovanni Ristori
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Franco Giubilei
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Marco Salvetti
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Francesco Orzi
- Department of Neurosciences Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Charles R G Guttmann
- Harvard Medical School, Center for Neurological Imaging, Brigham and Women's Hospital, Boston, MA, United States
| | - Michele Cavallari
- Harvard Medical School, Center for Neurological Imaging, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
44
|
Wang X, Ding D, Zhao Q, Liang X, Peng L, Zhao X, Xi Q, Min Z, Wang W, Xu X, Guo Q, Wang PJ. Brain hemodynamic changes in amnestic mild cognitive impairment measured by pulsed arterial spin labeling. Aging (Albany NY) 2020; 12:4348-4356. [PMID: 32167487 PMCID: PMC7093201 DOI: 10.18632/aging.102888] [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: 09/05/2019] [Accepted: 02/22/2020] [Indexed: 01/10/2023]
Abstract
We used pulsed arterial spin labeling (PASL) to investigate differences in cerebral blood flow (CBF) between 26 patients with amnestic mild cognitive impairment (aMCI) and 27 controls with normal cognition (NC). Hypoperfusion was observed in the right temporal pole of the middle temporal gyrus and the right inferior temporal gyrus in the aMCI compared with NC group. Interestingly, hyperperfusion was observed in the left temporal pole of the middle temporal gyrus, left superior temporal gyrus, bilateral precuneus, postcentral gyrus, right inferior parietal lobule, and right angular gyrus in the aMCI group, which likely resulted from a compensatory mechanism to maintain advanced neural activities. We found that mean CBF in the right inferior temporal gyrus, precuneus, and postcentral gyrus was positively correlated with cognitive ability in the aMCI but not NC group. Collectively, our data indicate that PASL is a useful noninvasive technique for monitoring changes in CBF and predicting cognitive decline in aMCI.
Collapse
Affiliation(s)
- Xiangbin Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Ding Ding
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Qianhua Zhao
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Xiaoniu Liang
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Ling Peng
- Department of Radiology, Shanghai Liqun Hospital, Shanghai 200333, PR China
| | - Xiaohu Zhao
- Department of Radiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Qian Xi
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Zhang Min
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Wei Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Xiaowen Xu
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Qihao Guo
- Department of Geriatrics, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai 200233, PR China
| | - Pei-Jun Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| |
Collapse
|
45
|
Klohs J. An Integrated View on Vascular Dysfunction in Alzheimer's Disease. NEURODEGENER DIS 2020; 19:109-127. [PMID: 32062666 DOI: 10.1159/000505625] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cerebrovascular disease is a common comorbidity in patients with Alzheimer's disease (AD). It is believed to contribute additively to the cognitive impairment and to lower the threshold for the development of dementia. However, accumulating evidence suggests that dysfunction of the cerebral vasculature and AD neuropathology interact in multiple ways. Vascular processes even proceed AD neuropathology, implicating a causal role in the etiology of AD. Thus, the review aims to provide an integrated view on vascular dysfunction in AD. SUMMARY In AD, the cerebral vasculature undergoes pronounced cellular, morphological and structural changes, which alters regulation of blood flow, vascular fluid dynamics and vessel integrity. Stiffening of central blood vessels lead to transmission of excessive pulsatile energy to the brain microvasculature, causing end-organ damage. Moreover, a dysregulated hemostasis and chronic vascular inflammation further impede vascular function, where its mediators interact synergistically. Changes of the cerebral vasculature are triggered and driven by systemic vascular abnormalities that are part of aging, and which can be accelerated and aggravated by cardiovascular diseases. Key Messages: In AD, the cerebral vasculature is the locus where multiple pathogenic processes converge and contribute to cognitive impairment. Understanding the molecular mechanism and pathophysiology of vascular dysfunction in AD and use of vascular blood-based and imaging biomarker in clinical studies may hold promise for future prevention and therapy of the disease.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland, .,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland,
| |
Collapse
|
46
|
Greenberg SM, Bacskai BJ, Hernandez-Guillamon M, Pruzin J, Sperling R, van Veluw SJ. Cerebral amyloid angiopathy and Alzheimer disease - one peptide, two pathways. Nat Rev Neurol 2020; 16:30-42. [PMID: 31827267 PMCID: PMC7268202 DOI: 10.1038/s41582-019-0281-2] [Citation(s) in RCA: 421] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2019] [Indexed: 12/22/2022]
Abstract
The shared role of amyloid-β (Aβ) deposition in cerebral amyloid angiopathy (CAA) and Alzheimer disease (AD) is arguably the clearest instance of crosstalk between neurodegenerative and cerebrovascular processes. The pathogenic pathways of CAA and AD intersect at the levels of Aβ generation, its circulation within the interstitial fluid and perivascular drainage pathways and its brain clearance, but diverge in their mechanisms of brain injury and disease presentation. Here, we review the evidence for and the pathogenic implications of interactions between CAA and AD. Both pathologies seem to be driven by impaired Aβ clearance, creating conditions for a self-reinforcing cycle of increased vascular Aβ, reduced perivascular clearance and further CAA and AD progression. Despite the close relationship between vascular and plaque Aβ deposition, several factors favour one or the other, such as the carboxy-terminal site of the peptide and specific co-deposited proteins. Amyloid-related imaging abnormalities that have been seen in trials of anti-Aβ immunotherapy are another probable intersection between CAA and AD, representing overload of perivascular clearance pathways and the effects of removing Aβ from CAA-positive vessels. The intersections between CAA and AD point to a crucial role for improving vascular function in the treatment of both diseases and indicate the next steps necessary for identifying therapies.
Collapse
Affiliation(s)
- Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Brian J Bacskai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mar Hernandez-Guillamon
- Neurovascular Research Laboratory, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jeremy Pruzin
- Center for Alzheimer Research and Treatment, Brigham & Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa Sperling
- Center for Alzheimer Research and Treatment, Brigham & Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Susanne J van Veluw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Yamazaki Y, Shinohara M, Yamazaki A, Ren Y, Asmann YW, Kanekiyo T, Bu G. ApoE (Apolipoprotein E) in Brain Pericytes Regulates Endothelial Function in an Isoform-Dependent Manner by Modulating Basement Membrane Components. Arterioscler Thromb Vasc Biol 2020; 40:128-144. [PMID: 31665905 PMCID: PMC7007705 DOI: 10.1161/atvbaha.119.313169] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/17/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The ε4 allele of the APOE gene (APOE4) is the strongest genetic risk factor for Alzheimer disease when compared with the common ε3 allele. Although there has been significant progress in understanding how apoE4 (apolipoprotein E4) drives amyloid pathology, its effects on amyloid-independent pathways, in particular cerebrovascular integrity and function, are less clear. Approach and Results: Here, we show that brain pericytes, the mural cells of the capillary walls, differentially modulate endothelial cell phenotype in an apoE isoform-dependent manner. Extracellular matrix protein induction, tube-like structure formation, and barrier formation were lower with endothelial cells cocultured with pericytes isolated from apoE4-targeted replacement (TR) mice compared with those from apoE3-TR mice. Importantly, aged apoE4-targeted replacement mice had decreased extracellular matrix protein expression and increased plasma protein leakages compared with apoE3-TR mice. CONCLUSIONS ApoE4 impairs pericyte-mediated basement membrane formation, potentially contributing to the cerebrovascular effects of apoE4.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | | | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - Yingxue Ren
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224
| | - Yan W. Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224
| | | | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| |
Collapse
|
48
|
Dewing JM, Carare RO, Lotery AJ, Ratnayaka JA. The Diverse Roles of TIMP-3: Insights into Degenerative Diseases of the Senescent Retina and Brain. Cells 2019; 9:cells9010039. [PMID: 31877820 PMCID: PMC7017234 DOI: 10.3390/cells9010039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Tissue inhibitor of metalloproteinase-3 (TIMP-3) is a component of the extracellular environment, where it mediates diverse processes including matrix regulation/turnover, inflammation and angiogenesis. Rare TIMP-3 risk alleles and mutations are directly linked with retinopathies such as age-related macular degeneration (AMD) and Sorsby fundus dystrophy, and potentially, through indirect mechanisms, with Alzheimer's disease. Insights into TIMP-3 activities may be gleaned from studying Sorsby-linked mutations. However, recent findings do not fully support the prevailing hypothesis that a gain of function through the dimerisation of mutated TIMP-3 is responsible for retinopathy. Findings from Alzheimer's patients suggest a hitherto poorly studied relationship between TIMP-3 and the Alzheimer's-linked amyloid-beta (A) proteins that warrant further scrutiny. This may also have implications for understanding AMD as aged/diseased retinae contain high levels of A. Findings from TIMP-3 knockout and mutant knock-in mice have not led to new treatments, particularly as the latter does not satisfactorily recapitulate the Sorsby phenotype. However, recent advances in stem cell and in vitro approaches offer novel insights into understanding TIMP-3 pathology in the retina-brain axis, which has so far not been collectively examined. We propose that TIMP-3 activities could extend beyond its hitherto supposed functions to cause age-related changes and disease in these organs.
Collapse
Affiliation(s)
- Jennifer M. Dewing
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
| | - Roxana O. Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
| | - Andrew J. Lotery
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
- Eye Unit, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - J. Arjuna Ratnayaka
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, MP806, Tremona Road, Southampton SO16 6YD, UK; (J.M.D.); (R.O.C.); (A.J.L.)
- Correspondence: ; Tel.: +44-238120-8183
| |
Collapse
|
49
|
Boese AC, Hamblin MH, Lee JP. Neural stem cell therapy for neurovascular injury in Alzheimer's disease. Exp Neurol 2019; 324:113112. [PMID: 31730762 DOI: 10.1016/j.expneurol.2019.113112] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by progressive neurodegeneration leading to severe cognitive decline and eventual death. AD pathophysiology is complex, but neurotoxic accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau are believed to be main drivers of neurodegeneration in AD. The formation and deposition of Aβ plaques occurs in the brain parenchyma as well as in the cerebral vasculature. Thus, proper blood-brain barrier (BBB) and cerebrovascular functioning are crucial for clearance of Aβ from the brain, and neurovascular dysfunction may be a critical component of AD development. Further, neuroinflammation and dysfunction of angiogenesis, neurogenesis, and neurorestorative capabilities play a role in AD pathophysiology. Currently, there is no effective treatment to prevent or restore loss of brain tissue and cognitive decline in patients with AD. Based on multifactorial and complex pathophysiological cascades in multiple Alzheimer's disease stages, effective AD therapies need to focus on targeting early AD pathology and preserving cerebrovascular function. Neural stem cells (NSCs) participate extensively in mammalian brain homeostasis and repair and exhibit pleiotropic intrinsic properties that likely make them attractive candidates for the treatment of AD. In the review, we summarize the current advances in knowledge regarding neurovascular aspects of AD-related neurodegeneration and discuss multiple actions of NSCs from preclinical studies of AD to evaluate their potential for future clinical treatment of AD.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
50
|
Reed MJ, Damodarasamy M, Banks WA. The extracellular matrix of the blood-brain barrier: structural and functional roles in health, aging, and Alzheimer's disease. Tissue Barriers 2019; 7:1651157. [PMID: 31505997 DOI: 10.1080/21688370.2019.1651157] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is increasing interest in defining the location, content, and role of extracellular matrix (ECM) components in brain structure and function during development, aging, injury, and neurodegeneration. Studies in vivo confirm brain ECM has a dynamic composition with constitutive and induced alterations that impact subsequent cell-cell and cell-matrix interactions. Moreover, it is clear that for any given ECM component, the brain region, and cell type within that location, determines the direction, magnitude, and composition of those changes. This review will examine the ECM at the neurovascular unit (NVU) and the blood-brain barrier (BBB) within the NVU. The discussion will begin at the glycocalyx ECM on the luminal surface of the vasculature, and progress to the abluminal side with a focus on changes in basement membrane ECM during aging and neurodegeneration.
Collapse
Affiliation(s)
- May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA.,VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA
| |
Collapse
|