1
|
Nyúl-Tóth Á, Patai R, Csiszar A, Ungvari A, Gulej R, Mukli P, Yabluchanskiy A, Benyo Z, Sotonyi P, Prodan CI, Liotta EM, Toth P, Elahi F, Barsi P, Maurovich-Horvat P, Sorond FA, Tarantini S, Ungvari Z. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. GeroScience 2024; 46:6511-6536. [PMID: 38831182 PMCID: PMC11494622 DOI: 10.1007/s11357-024-01194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Aging plays a pivotal role in the pathogenesis of cerebral small vessel disease (CSVD), contributing to the onset and progression of vascular cognitive impairment and dementia (VCID). In older adults, CSVD often leads to significant pathological outcomes, including blood-brain barrier (BBB) disruption, which in turn triggers neuroinflammation and white matter damage. This damage is frequently observed as white matter hyperintensities (WMHs) in neuroimaging studies. There is mounting evidence that older adults with atherosclerotic vascular diseases, such as peripheral artery disease, ischemic heart disease, and carotid artery stenosis, face a heightened risk of developing CSVD and VCID. This review explores the complex relationship between peripheral atherosclerosis, the pathogenesis of CSVD, and BBB disruption. It explores the continuum of vascular aging, emphasizing the shared pathomechanisms that underlie atherosclerosis in large arteries and BBB disruption in the cerebral microcirculation, exacerbating both CSVD and VCID. By reviewing current evidence, this paper discusses the impact of endothelial dysfunction, cellular senescence, inflammation, and oxidative stress on vascular and neurovascular health. This review aims to enhance understanding of these complex interactions and advocate for integrated approaches to manage vascular health, thereby mitigating the risk and progression of CSVD and VCID.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN, Semmelweis University, 1094, Budapest, Hungary
| | - Peter Sotonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Eric M Liotta
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Peter Toth
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Neurotrauma Research Group, Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, University of Pecs, Pecs, Hungary
| | - Fanny Elahi
- Departments of Neurology and Neuroscience Ronald M. Loeb Center for Alzheimer's Disease Friedman Brain Institute Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Péter Barsi
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Pál Maurovich-Horvat
- ELKH-SE Cardiovascular Imaging Research Group, Department of Radiology, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Farzaneh A Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Department of Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| |
Collapse
|
2
|
Palmer JA, Kaufman CS, Whitaker-Hilbig AA, Billinger SA. APOE4 carriers display loss of anticipatory cerebral vascular regulation over AD progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.11.24315344. [PMID: 39417136 PMCID: PMC11482999 DOI: 10.1101/2024.10.11.24315344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Maintenance of cerebral blood flow during orthostasis is impaired with aging and associated with cognitive decline, but the effect of Apolipoprotein 4-allele (APOE4) is unknown. METHODS Older adults (n=108) (APOE4 carriers, n=47; noncarriers, n=61) diagnosed as cognitively-normal (NC), MCI, or AD participated. Middle cerebral artery blood velocity (MCAv), assessed using Transcranial Doppler ultrasound, and beat-to-beat mean arterial blood pressure (MAP) were continuously recorded during a sit-to-stand transition. Anticipatory and orthostatic-induced MCAv and MAP responses were compared between genotypes and across disease progression. RESULTS Cognitively-normal APOE4 carriers showed greater anticipatory MCAv increase, greater MCAv decrease with orthostasis, and shorter latency of peripheral MAP responses to orthostasis compared to noncarriers. MCAv and MAP responses were delayed and attenuated across the APOE4 disease progression, with no differences between genotypes in MCI and AD. DISCUSSION APOE4 carriers and noncarriers present with distinct phenotypes of cerebral vascular dysfunction during hemodynamic orthostatic challenge. Unique cerebral and peripheral vascular compensation observed in APOE4 carriers may be lost as AD progresses.
Collapse
Affiliation(s)
- Jacqueline A. Palmer
- Division of Physical Therapy and Rehabilitation Science, University of Minnesota Medical School, , Minneapolis, MN, United States of America
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| | - Carolyn S. Kaufman
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, United States of America
- Department of Internal Medicine, Stanford Health Care, Stanford University, Palo Alto, CA, United States of America
| | - Alicen A. Whitaker-Hilbig
- Department of Physical Medicine and Rehabilitation, Medical College of Wisconsin, 123 Milwaukee Way, Milwaukee, WI, United States of America
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Sandra A. Billinger
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States of America
- University of Kansas Alzheimer’s Disease Research Center, Fairway, KS, United States of America
| |
Collapse
|
3
|
Lim XR, Abd-Alhaseeb MM, Ippolito M, Koide M, Senatore AJ, Plante C, Hariharan A, Weir N, Longden TA, Laprade KA, Stafford JM, Ziemens D, Schwaninger M, Wenzel J, Postnov DD, Harraz OF. Endothelial Piezo1 channel mediates mechano-feedback control of brain blood flow. Nat Commun 2024; 15:8686. [PMID: 39375369 PMCID: PMC11458797 DOI: 10.1038/s41467-024-52969-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
Hyperemia in response to neural activity is essential for brain health. A hyperemic response delivers O2 and nutrients, clears metabolic waste, and concomitantly exposes cerebrovascular endothelial cells to hemodynamic forces. While neurovascular research has primarily centered on the front end of hyperemia-neuronal activity-to-vascular response-the mechanical consequences of hyperemia have gone largely unexplored. Piezo1 is an endothelial mechanosensor that senses hyperemia-associated forces. Using genetic mouse models and pharmacologic approaches to manipulate endothelial Piezo1 function, we evaluated its role in blood flow control and whether it impacts cognition. We provide evidence of a built-in brake system that sculpts hyperemia, and specifically show that Piezo1 activation triggers a mechano-feedback system that promotes blood flow recovery to baseline. Further, genetic Piezo1 modification led to deficits in complementary memory tasks. Collectively, our findings establish a role for endothelial Piezo1 in cerebral blood flow regulation and a role in its behavioral sequelae.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Michael Ippolito
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Masayo Koide
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Amanda J Senatore
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Curtis Plante
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA
| | - Ashwini Hariharan
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nick Weir
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Thomas A Longden
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Laboratory of Neurovascular Interactions, Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Kathryn A Laprade
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - James M Stafford
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Dorothea Ziemens
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- German Research Centre for Cardiovascular Research (DZHK), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Dmitry D Postnov
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, 8200, Denmark
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA.
- Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, VT, USA.
| |
Collapse
|
4
|
Anfray A, Schaeffer S, Hattori Y, Santisteban MM, Casey N, Wang G, Strickland M, Zhou P, Holtzman DM, Anrather J, Park L, Iadecola C. A cell-autonomous role for border-associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. Nat Neurosci 2024:10.1038/s41593-024-01757-6. [PMID: 39294490 DOI: 10.1038/s41593-024-01757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Apolipoprotein E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4, we report that border-associated macrophages (BAMs), myeloid cells closely apposed to neocortical microvessels, are both sources and effectors of ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAMs is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell-autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
- Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Samantha Schaeffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Monica M Santisteban
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Strickland
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
6
|
Aamand R, Rasmussen PM, Andersen KS, de Paoli S, Weitzberg E, Christiansen M, Lund TE, Østergaard L. Cerebral microvascular changes in healthy carriers of the APOE-ɛ4 Alzheimer's disease risk gene. PNAS NEXUS 2024; 3:pgae369. [PMID: 39253395 PMCID: PMC11382292 DOI: 10.1093/pnasnexus/pgae369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
APOE-ɛ4 is a genetic risk factor for Alzheimer's disease (AD). AD is associated with reduced cerebral blood flow (CBF) and with microvascular changes that limit the transport of oxygen from blood into brain tissue: reduced microvascular cerebral blood volume and high relative transit time heterogeneity (RTH). Healthy APOE-ɛ4 carriers reveal brain regions with elevated CBF compared with carriers of the common ɛ3 allele. Such asymptomatic hyperemia may reflect microvascular dysfunction: a vascular disease entity characterized by suboptimal tissue oxygen uptake, rather than limited blood flow per se. Here, we used perfusion MRI to show that elevated regional CBF is accompanied by reduced capillary blood volume in healthy APOE-ɛ4 carriers (carriers) aged 30-70 years compared with similarly aged APOE-ɛ3 carriers (noncarriers). Younger carriers have elevated hippocampal RTH and more extreme RTH values throughout both white matter (WM) and cortical gray matter (GM) compared with noncarriers. Older carriers have reduced WM CBF and more extreme GM RTH values than noncarriers. Across all groups, lower WM and hippocampal RTH correlate with higher educational attainment, which is associated with lower AD risk. Three days of dietary nitrate supplementation increased carriers' WM CBF but caused older carriers to score worse on two of six aggregate neuropsychological scores. The intervention improved late recall in younger carriers and in noncarriers. The APOE-ɛ4 gene is associated with microvascular changes that may impair tissue oxygen extraction. We speculate that vascular risk factor control is particularly important for APOE-ɛ4 carriers' healthy aging.
Collapse
Affiliation(s)
- Rasmus Aamand
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Peter M Rasmussen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Katrine Schilling Andersen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Stine de Paoli
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Torben E Lund
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
- Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
7
|
Diany R, Gagliano Taliun SA. Systematic Review and Phenome-Wide Scans of Genetic Associations with Vascular Cognitive Impairment. Adv Biol (Weinh) 2024; 8:e2300692. [PMID: 38935518 DOI: 10.1002/adbi.202300692] [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: 12/16/2023] [Revised: 03/12/2024] [Indexed: 06/29/2024]
Abstract
Vascular cognitive impairment (VCI) is a heterogenous form of cognitive impairment that results from cerebrovascular disease. It is a result of both genetic and non-genetic factors. Although much research has been conducted on the genetic contributors to other forms of cognitive impairment (e.g. Alzheimer's disease), knowledge is lacking on the genetic factors associated with VCI. A better understanding of the genetics of VCI will be critical for prevention and treatment. To begin to fill this gap, the genetic contributors are reviewed with VCI from the literature. Phenome-wide scans of the identified genes are conducted and genetic variants identified in the review in large-scale resources displaying genetic variant-trait association information. Gene set are also carried out enrichment analysis using the genes identified from the review. Thirty one articles are identified meeting the search criteria and filters, from which 107 unique protein-coding genes are noted related to VCI. The phenome-wide scans and gene set enrichment analysis identify pathways associated with a diverse set of biological systems. This results indicate that genes with evidence of involvement in VCI are involved in a diverse set of biological functions. This information can facilitate downstream research to better dissect possible shared biological mechanisms for future therapies.
Collapse
Affiliation(s)
- Rime Diany
- Faculty of Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3C 3J7, Canada
| | - Sarah A Gagliano Taliun
- Department of Medicine & Department of Neurosciences, Faculty of Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3C 3J7, Canada
- Montreal Heart Institute, 5000 rue Bélanger, Montréal, Québec, H1T 1C8, Canada
| |
Collapse
|
8
|
van Heuvelen MJG, van der Lei MB, Alferink PM, Roemers P, van der Zee EA. Cognitive deficits in human ApoE4 knock-in mice: A systematic review and meta-analysis. Behav Brain Res 2024; 471:115123. [PMID: 38972485 DOI: 10.1016/j.bbr.2024.115123] [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: 02/21/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
Apolipoprotein-E4 (ApoE4) is an important genetic risk factor for Alzheimer's disease. The development of targeted-replacement human ApoE knock-in mice facilitates research into mechanisms by which ApoE4 affects the brain. We performed meta-analyses and meta-regression analyses to examine differences in cognitive performance between ApoE4 and ApoE3 mice. We included 61 studies in which at least one of the following tests was assessed: Morris Water Maze (MWM), novel object location (NL), novel object recognition (NO) and Fear Conditioning (FC) test. ApoE4 vs. ApoE3 mice performed significantly worse on the MWM (several outcomes, 0.17 ≤ g ≤ 0.60), NO (exploration, g=0.33; index, g=0.44) and FC (contextual, g=0.49). ApoE4 vs. ApoE3 differences were not systematically related to sex or age. We conclude that ApoE4 knock-in mice in a non-AD condition show some, but limited cognitive deficits, regardless of sex and age. These effects suggest an intrinsic vulnerability in ApoE4 mice that may become more pronounced under additional brain load, as seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Marieke J G van Heuvelen
- Department of Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, Groningen 9713AV, the Netherlands.
| | - Mathijs B van der Lei
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands; Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, Edegem 2650, Belgium.
| | - Pien M Alferink
- Department of Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, Groningen 9713AV, the Netherlands.
| | - Peter Roemers
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands.
| | - Eddy A van der Zee
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands.
| |
Collapse
|
9
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [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] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
10
|
Kakae M, Kawashita A, Onogi H, Nakagawa T, Shirakawa H. Bilateral Common Carotid Artery Stenosis in Mice: A Model of Chronic Cerebral Hypoperfusion-Induced Vascular Cognitive Impairment. Bio Protoc 2024; 14:e5022. [PMID: 39007157 PMCID: PMC11238111 DOI: 10.21769/bioprotoc.5022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/30/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Vascular cognitive impairment (VCI) is a syndrome defined as cognitive decline caused by vascular disease and is associated with various types of dementia. Chronic cerebral hypoperfusion (CCH) is one of the major contributors to VCI. Among the various rodent models used to study CCH-induced VCI, we have found the mouse bilateral common carotid artery stenosis (BCAS) model to be highly suitable. Here, we introduce the BCAS model of C57BL/6J mice generated using microcoils with an internal diameter of 0.18 mm. To produce the mouse BCAS model, the bilateral common carotid arteries are isolated from the adhering tissues and vagus nerves and twined around the microcoils. This model shows cognitive impairment and white matter lesions preceding neuronal dysfunction around postoperative day 28, which is similar to the human clinical picture. Overall, the mouse BCAS model will continue to be useful in studying CCH-induced VCI. Key features • This mouse BCAS model requires approximately 4 weeks to show phenotypes such as cognitive impairment and white matter injury.
Collapse
Affiliation(s)
- Masashi Kakae
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Department of Clinical Pharmacology and Pharmacotherapy, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Ayaka Kawashita
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Haruya Onogi
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Pharmacotherapy, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Kapoor A, Dutt S, Alitin JPM, Sible IJ, Marshall A, Shenasa F, Engstrom AC, Gaubert A, Shao X, Bradford DR, Rodgers K, Mather M, Wang DJJ, Nation DA. Older adults with reduced cerebrovascular reactivity exhibit high white matter hyperintensity burden. Neurobiol Aging 2024; 139:5-10. [PMID: 38579393 DOI: 10.1016/j.neurobiolaging.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Cerebrovascular reactivity (CVR) deficits may contribute to small vessel disease, such as white matter hyperintensities (WMH). Moreover, apolipoprotein-e4 (APOE4) carriers at genetic risk for Alzheimer's disease exhibit cerebrovascular dysfunction relative to non-carriers. We examined whether older adults, and APOE4 carriers specifically, with diminished CVR would exhibit higher WMH burden. Independently living older adults (N = 125, mean age = 69.2 years; SD = 7.6; 31.2% male) free of dementia or clinical stroke underwent brain MRI to quantify cerebral perfusion during CVR to hypercapnia and hypocapnia and determine WMH volume. Adjusting for age, sex and intracranial volume, hierarchical regression analysis revealed a significant association between whole brain CVR to hypercapnia and WMH overall [B = -.02, 95% CI (-.04, -.008), p =.003] and in APOE4 carriers [B = -.03, 95% CI (-.06, -.009), p =.009]. Findings suggest deficits in cerebral vasodilatory capacity are associated with WMH burden in older adults and future studies are warranted to further delineate the effect of APOE4 on precipitating WMH.
Collapse
Affiliation(s)
- Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - John Paul M Alitin
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Aimée Gaubert
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Xingfeng Shao
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - David Robert Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Mara Mather
- University of Southern California Leonard Davis School of Gerontology, USA
| | - Danny J J Wang
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- University of Southern California Leonard Davis School of Gerontology, USA; Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California Keck School of Medicine, USA.
| |
Collapse
|
12
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [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: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
13
|
Wang X, Wen D, Xia F, Fang M, Zheng J, You C, Ma L. Single-Cell Transcriptomics Revealed White Matter Repair Following Subarachnoid Hemorrhage. Transl Stroke Res 2024:10.1007/s12975-024-01265-6. [PMID: 38861152 DOI: 10.1007/s12975-024-01265-6] [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: 05/02/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Existing research indicates the potential for white matter injury repair during the subacute phase following subarachnoid hemorrhage (SAH). However, elucidating the role of brain cell subpopulations in the acute and subacute phases of SAH pathogenesis remains challenging due to the cellular heterogeneity of the central nervous system. In this study, single-cell RNA sequencing was conducted on SAH model mice to delineate distinct cell populations. Gene Set Enrichment Analysis was performed to identify involved pathways, and cellular interactions were explored using the CellChat package in R software. Validation of the findings involved a comprehensive approach, including magnetic resonance imaging, immunofluorescence double staining, and Western blot analyses. This study identified ten major brain clusters with cell type-specific gene expression patterns. Notably, we observed infiltration and clonal expansion of reparative microglia in white matter-enriched regions during the subacute stage after SAH. Additionally, microglia-associated pleiotrophin (PTN) was identified as having a role in mediating the regulation of oligodendrocyte precursor cells (OPCs) in SAH model mice, implicating the activation of the mTOR signaling pathway. These findings emphasize the vital role of microglia-OPC interactions might occur via the PTN pathway, potentially contributing to white matter repair during the subacute phase after SAH. Our analysis revealed precise transcriptional changes in the acute and subacute phases after SAH, offering insights into the mechanism of SAH and for the development of drugs that target-specific cell subtypes.
Collapse
Affiliation(s)
- Xing Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dingke Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mei Fang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jun Zheng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Centre, Sichuan University, Chengdu, Sichuan, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
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
|
15
|
Wang H, Shi L, Luo S, Luo Y, Xu C, Qiu G, Guo Q, Chen C, Lu T, Liu K, Zhu F. Associations of apolipoprotein E ε4 allele, regional cerebral blood flow, and serum liver function markers in patients with cognitive impairment. Front Neurol 2024; 15:1345705. [PMID: 38628697 PMCID: PMC11018914 DOI: 10.3389/fneur.2024.1345705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction The ε4 allele of the apolipoprotein E gene (APOE4) is expressed abundantly in both the brain and peripheral circulation as a genetic risk factor for Alzheimer's disease (AD). Cerebral blood flow (CBF) dysfunction is an essential feature of AD, and the liver plays an important role in the pathogenesis of dementia. However, the associations of APOE4 with CBF and liver function markers in patients with cognitive impairment remains unclear. We aimed to evaluate the associations of APOE4 with CBF measured by arterial spin labeling (ASL) magnetic resonance imaging (MRI) and serum liver function markers in participants who were diagnosed with cognitive impairment. Methods Fourteen participants with AD and sixteen with amnestic mild cognitive impairment (MCI) were recruited. In addition to providing comprehensive clinical information, all patients underwent laboratory tests and MRI. All participants were divided into carriers and noncarriers of the ε4 allele, and T-tests and Mann-Whitney U tests were used to observe the differences between APOE4 carriers and noncarriers in CBF and liver function markers. Results Regarding regional cerebral blood flow (rCBF), APOE4 carriers showed hyperperfusion in the bilateral occipital cortex, bilateral thalamus, and left precuneus and hypoperfusion in the right lateral temporal cortex when compared with noncarriers. Regarding serum liver function markers, bilirubin levels (including total, direct, and indirect) were lower in APOE4 carriers than in noncarriers. Conclusion APOE4 exerts a strong effect on CBF dysfunction by inheritance, representing a risk factor for AD. APOE4 may be related to bilirubin metabolism, potentially providing specific neural targets for the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Hao Wang
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lin Shi
- BrainNow Research Institute, Guangdong, China
| | - Shimei Luo
- Department of Nuclear Magnetic Resonance, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Yishan Luo
- BrainNow Research Institute, Guangdong, China
| | - Chunyan Xu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Guozhen Qiu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Qiwen Guo
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Chunchun Chen
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Taikun Lu
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kangding Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| |
Collapse
|
16
|
Reeve EH, Barnes JN, Moir ME, Walker AE. Impact of arterial stiffness on cerebrovascular function: a review of evidence from humans and preclincal models. Am J Physiol Heart Circ Physiol 2024; 326:H689-H704. [PMID: 38214904 PMCID: PMC11221809 DOI: 10.1152/ajpheart.00592.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
With advancing age, the cerebral vasculature becomes dysfunctional, and this dysfunction is associated with cognitive decline. However, the initiating cause of these age-related cerebrovascular impairments remains incompletely understood. A characteristic feature of the aging vasculature is the increase in stiffness of the large elastic arteries. This increase in arterial stiffness is associated with elevated pulse pressure and blood flow pulsatility in the cerebral vasculature. Evidence from both humans and rodents supports that increases in large elastic artery stiffness are associated with cerebrovascular impairments. These impacts on cerebrovascular function are wide-ranging and include reductions in global and regional cerebral blood flow, cerebral small vessel disease, endothelial cell dysfunction, and impaired perivascular clearance. Furthermore, recent findings suggest that the relationship between arterial stiffness and cerebrovascular function may be influenced by genetics, specifically APOE and NOTCH genotypes. Given the strength of the evidence that age-related increases in arterial stiffness have deleterious impacts on the brain, interventions that target arterial stiffness are needed. The purpose of this review is to summarize the evidence from human and rodent studies, supporting the role of increased arterial stiffness in age-related cerebrovascular impairments.
Collapse
Affiliation(s)
- Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jill N Barnes
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - M Erin Moir
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
17
|
Stamenkovic S, Schmid F, Weitermann N, Takasaki K, Bonney SK, Sosa MJ, Li Y, Bennett HC, Kim Y, Waters J, Shih AY. Impaired drainage through capillary-venous networks contributes to age-related white matter loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.11.579849. [PMID: 38405879 PMCID: PMC10888936 DOI: 10.1101/2024.02.11.579849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The gradual loss of cerebral white matter contributes to cognitive decline during aging. However, microvascular networks that support the metabolic demands of white matter remain poorly defined. We used in vivo deep multi-photon imaging to characterize microvascular networks that perfuse cortical layer 6 and corpus callosum, a highly studied region of white matter in the mouse brain. We show that these deep tissues are exclusively drained by sparse and wide-reaching venules, termed principal cortical venules, which mirror vascular architecture at the human cortical-U fiber interface. During aging, capillary networks draining into deep branches of principal cortical venules are selectively constricted, reduced in density, and diminished in pericyte numbers. This causes hypo-perfusion in deep tissues, and correlates with gliosis and demyelination, whereas superficial tissues become relatively hyper-perfused. Thus, age-related impairment of capillary-venular drainage is a key vascular deficit that contributes to the unique vulnerability of cerebral white matter during brain aging.
Collapse
|
18
|
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
|
19
|
Bonnar O, Shaw K, Anderle S, Grijseels DM, Clarke D, Bell L, King SL, Hall CN. APOE4 expression confers a mild, persistent reduction in neurovascular function in the visual cortex and hippocampus of awake mice. J Cereb Blood Flow Metab 2023; 43:1826-1841. [PMID: 37350319 PMCID: PMC10676141 DOI: 10.1177/0271678x231172842] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 06/24/2023]
Abstract
Vascular factors are known to be early and important players in Alzheimer's disease (AD) development, however the role of the ε4 allele of the Apolipoprotein (APOE) gene (a risk factor for developing AD) remains unclear. APOE4 genotype is associated with early and severe neocortical vascular deficits in anaesthetised mice, but in humans, vascular and cognitive dysfunction are focused on the hippocampal formation and appear later. How APOE4 might interact with the vasculature to confer AD risk during the preclinical phase represents a gap in existing knowledge. To avoid potential confounds of anaesthesia and to explore regions most relevant for human disease, we studied the visual cortex and hippocampus of awake APOE3 and APOE4-TR mice using 2-photon microscopy of neurons and blood vessels. We found mild vascular deficits: vascular density and functional hyperaemia were unaffected in APOE4 mice, and neuronal or vascular function did not decrease up to late middle-age. Instead, vascular responsiveness was lower, arteriole vasomotion was reduced and neuronal calcium signals during visual stimulation were increased. This suggests that, alone, APOE4 expression is not catastrophic but stably alters neurovascular physiology. We suggest this state makes APOE4 carriers more sensitive to subsequent insults such as injury or beta amyloid accumulation.
Collapse
Affiliation(s)
| | | | - Silvia Anderle
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Dori M Grijseels
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Devin Clarke
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Laura Bell
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Sarah L King
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Catherine N Hall
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| |
Collapse
|
20
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
21
|
Wang Z, Nie X, Gao F, Tang Y, Ma Y, Zhang Y, Gao Y, Yang C, Ding J, Wang X. Increasing brain N-acetylneuraminic acid alleviates hydrocephalus-induced neurological deficits. CNS Neurosci Ther 2023; 29:3183-3198. [PMID: 37222223 PMCID: PMC10580356 DOI: 10.1111/cns.14253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/27/2023] [Accepted: 04/01/2023] [Indexed: 05/25/2023] Open
Abstract
AIMS This metabolomic study aimed to evaluate the role of N-acetylneuraminic acid (Neu5Ac) in the neurological deficits of normal pressure hydrocephalus (NPH) and its potential therapeutic effect. METHODS We analyzed the metabolic profiles of NPH using cerebrospinal fluid with multivariate and univariate statistical analyses in a set of 42 NPH patients and 38 controls. We further correlated the levels of differential metabolites with severity-related clinical parameters, including the normal pressure hydrocephalus grading scale (NPHGS). We then established kaolin-induced hydrocephalus in mice and treated them using N-acetylmannosamine (ManNAc), a precursor of Neu5Ac. We examined brain Neu5Ac, astrocyte polarization, demyelination, and neurobehavioral outcomes to explore its therapeutic effect. RESULTS Three metabolites were significantly altered in NPH patients. Only decreased Neu5Ac levels were correlated with NPHGS scores. Decreased brain Neu5Ac levels have been observed in hydrocephalic mice. Increasing brain Neu5Ac by ManNAc suppressed the activation of astrocytes and promoted their transition from A1 to A2 polarization. ManNAc also attenuated the periventricular white matter demyelination and improved neurobehavioral outcomes in hydrocephalic mice. CONCLUSION Increasing brain Neu5Ac improved the neurological outcomes associated with the regulation of astrocyte polarization and the suppression of demyelination in hydrocephalic mice, which may be a potential therapeutic strategy for NPH.
Collapse
Affiliation(s)
- Zhangyang Wang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xiaoqun Nie
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Fang Gao
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Yanmin Tang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yuanyuan Ma
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yiying Zhang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yanqin Gao
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Chen Yang
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Jing Ding
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xin Wang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
22
|
Uekawa K, Hattori Y, Ahn SJ, Seo J, Casey N, Anfray A, Zhou P, Luo W, Anrather J, Park L, Iadecola C. Border-associated macrophages promote cerebral amyloid angiopathy and cognitive impairment through vascular oxidative stress. Mol Neurodegener 2023; 18:73. [PMID: 37789345 PMCID: PMC10548599 DOI: 10.1186/s13024-023-00660-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 09/20/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a devastating condition common in patients with Alzheimer's disease but also observed in the general population. Vascular oxidative stress and neurovascular dysfunction have been implicated in CAA but the cellular source of reactive oxygen species (ROS) and related signaling mechanisms remain unclear. We tested the hypothesis that brain border-associated macrophages (BAM), yolk sac-derived myeloid cells closely apposed to parenchymal and leptomeningeal blood vessels, are the source of radicals through the Aβ-binding innate immunity receptor CD36, leading to neurovascular dysfunction, CAA, and cognitive impairment. METHODS Tg2576 mice and WT littermates were transplanted with CD36-/- or CD36+/+ bone marrow at 12-month of age and tested at 15 months. This approach enables the repopulation of perivascular and leptomeningeal compartments with CD36-/- BAM. Neurovascular function was tested in anesthetized mice equipped with a cranial window in which cerebral blood flow was monitored by laser-Doppler flowmetry. Amyloid pathology and cognitive function were also examined. RESULTS The increase in blood flow evoked by whisker stimulation (functional hyperemia) or by endothelial and smooth muscle vasoactivity was markedly attenuated in WT → Tg2576 chimeras but was fully restored in CD36-/- → Tg2576 chimeras, in which BAM ROS production was suppressed. CAA-associated Aβ1-40, but not Aβ1-42, was reduced in CD36-/- → Tg2576 chimeras. Similarly, CAA, but not parenchymal plaques, was reduced in CD36-/- → Tg2576 chimeras. These beneficial vascular effects were associated with cognitive improvement. Finally, CD36-/- mice were able to more efficiently clear exogenous Aβ1-40 injected into the neocortex or the striatum. CONCLUSIONS CD36 deletion in BAM suppresses ROS production and rescues the neurovascular dysfunction and damage induced by Aβ. CD36 deletion in BAM also reduced brain Aβ1-40 and ameliorated CAA without affecting parenchyma plaques. Lack of CD36 enhanced the vascular clearance of exogenous Aβ. Restoration of neurovascular function and attenuation of CAA resulted in a near complete rescue of cognitive function. Collectively, these data implicate brain BAM in the pathogenesis of CAA and raise the possibility that targeting BAM CD36 is beneficial in CAA and other conditions associated with vascular Aβ deposition and damage.
Collapse
Affiliation(s)
- Ken Uekawa
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Sung Ji Ahn
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - James Seo
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Wenjie Luo
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
23
|
Marottoli FM, Zhang H, Flores-Barrera E, Artur de la Villarmois E, Damen FC, Miguelez Fernández AM, Blesson HV, Chaudhary R, Nguyen AL, Nwokeji AE, Talati R, John AS, Madadakere K, Lutz SE, Cai K, Tseng KY, Tai LM. Endothelial Cell APOE3 Regulates Neurovascular, Neuronal, and Behavioral Function. Arterioscler Thromb Vasc Biol 2023; 43:1952-1966. [PMID: 37650329 PMCID: PMC10521805 DOI: 10.1161/atvbaha.123.319816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Specialized brain endothelial cells and human APOE3 are independently important for neurovascular function, yet whether APOE3 expression by endothelial cells contributes to brain function is currently unknown. In the present study, we determined whether the loss of endothelial cell APOE3 impacts brain vascular and neural function. METHODS We developed APOE3fl/fl/Cdh5(PAC)-CreERT2+/- (APOE3Cre+/-) and APOE3fl/fl/Cdh5(PAC)-CreERT2-/- (APOE3Cre-/-, control) mice and induced endothelial cell APOE3 knockdown with tamoxifen at ≈4 to 5 weeks of age. Neurovascular and neuronal function were evaluated by biochemistry, immunohistochemistry, behavioral testing, and electrophysiology at 9 months of age. RESULTS We found that the loss of endothelial APOE3 expression was sufficient to cause neurovascular dysfunction including higher permeability and lower vessel coverage in tandem with deficits in spatial memory and fear memory extinction and a disruption of cortical excitatory/inhibitory balance. CONCLUSIONS Our data collectively support the novel concept that endothelial APOE3 plays a critical role in the regulation of the neurovasculature, neural circuit function, and behavior.
Collapse
Affiliation(s)
- Felecia M. Marottoli
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hui Zhang
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Eden Flores-Barrera
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Emilce Artur de la Villarmois
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | | | - Anabel M.M. Miguelez Fernández
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hannah V. Blesson
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Rohan Chaudhary
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Anthony L. Nguyen
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Amanda E. Nwokeji
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ruju Talati
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ashwin S. John
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kushi Madadakere
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Sarah E. Lutz
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kejia Cai
- Radiology (F.C.D., K.C.), University of Illinois at Chicago
- Bioengineering (K.C.), University of Illinois at Chicago
| | - Kuei Y. Tseng
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Leon M. Tai
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| |
Collapse
|
24
|
Palmer JM, Huentelman M, Ryan L. More than just risk for Alzheimer's disease: APOE ε4's impact on the aging brain. Trends Neurosci 2023; 46:750-763. [PMID: 37460334 DOI: 10.1016/j.tins.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
The apolipoprotein ε4 (APOE ε4) allele is most commonly associated with increased risk for late-onset Alzheimer's disease (AD). However, recent longitudinal studies suggest that these risks are overestimated; most ε4 carriers will not develop dementia in their lifetime. In this article, we review new evidence regarding the impact of APOE ε4 on cognition among healthy older adults. We discuss emerging work from animal models suggesting that ε4 impacts brain structure and function in multiple ways that may lead to age-related cognitive impairment, independent from AD pathology. We discuss the importance of taking an individualized approach in future studies by incorporating biomarkers and neuroimaging methods that may better disentangle the phenotypic influences of APOE ε4 on the aging brain from prodromal AD pathology.
Collapse
Affiliation(s)
- Justin M Palmer
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Lee Ryan
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
25
|
Zhang Z, Kwapong WR, Cao L, Feng Z, Wu B, Liu J, Zhang S. APOE ε4 Gene Carriers Demonstrate Reduced Retinal Capillary Densities in Asymptomatic Older Adults. J Clin Med 2023; 12:5649. [PMID: 37685715 PMCID: PMC10488535 DOI: 10.3390/jcm12175649] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/06/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Early identification of Apolipoprotein E (APOE)-related microvascular pathology will help to study the microangiopathic contribution to Alzheimer's disease and provide a therapeutic target for early intervention. To evaluate the differences in retinal microvasculature parameters between APOE ε4 carriers and non-carriers, asymptomatic older adults aged ≥ 55 years underwent APOE ε4 genotype analysis, neuropsychological examination, and optical coherence tomography angiography (OCTA) imaging. One hundred sixty-three older adults were included in the data analysis. Participants were also defined as cognitively impaired (CI) and non-cognitively impaired (NCI) according to their MoCA scores and educational years. APOE ε4 carriers demonstrated reduced SVC (p = 0.023) compared to APOE ε4 non-carriers. Compared to NCI, CI participants showed reduced SVC density (p = 0.006). In the NCI group, no significant differences (p > 0.05) were observed in the microvascular densities between APOE ε4 carriers and non-carriers. In the CI group, APOE ε4 carriers displayed reduced microvascular densities compared to non-carriers (SVC, p = 0.006; DVC, p = 0.048). We showed that CI and APOE ε4 affect retinal microvasculature in older adults. Quantitative measures of the retinal microvasculature could serve as surrogates for brain microcirculation, providing an opportunity to study microvascular contributions to AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Junfeng Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
26
|
Rego S, Sanchez G, Da Mesquita S. Current views on meningeal lymphatics and immunity in aging and Alzheimer's disease. Mol Neurodegener 2023; 18:55. [PMID: 37580702 PMCID: PMC10424377 DOI: 10.1186/s13024-023-00645-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023] Open
Abstract
Alzheimer's disease (AD) is an aging-related form of dementia associated with the accumulation of pathological aggregates of amyloid beta and neurofibrillary tangles in the brain. These phenomena are accompanied by exacerbated inflammation and marked neuronal loss, which altogether contribute to accelerated cognitive decline. The multifactorial nature of AD, allied to our still limited knowledge of its etiology and pathophysiology, have lessened our capacity to develop effective treatments for AD patients. Over the last few decades, genome wide association studies and biomarker development, alongside mechanistic experiments involving animal models, have identified different immune components that play key roles in the modulation of brain pathology in AD, affecting its progression and severity. As we will relay in this review, much of the recent efforts have been directed to better understanding the role of brain innate immunity, and particularly of microglia. However, and despite the lack of diversity within brain resident immune cells, the brain border tissues, especially the meninges, harbour a considerable number of different types and subtypes of adaptive and innate immune cells. Alongside microglia, which have taken the centre stage as important players in AD research, there is new and exciting evidence pointing to adaptive immune cells, namely T and B cells found in the brain and its meninges, as important modulators of neuroinflammation and neuronal (dys)function in AD. Importantly, a genuine and functional lymphatic vascular network is present around the brain in the outermost meningeal layer, the dura. The meningeal lymphatics are directly connected to the peripheral lymphatic system in different mammalian species, including humans, and play a crucial role in preserving a "healthy" immune surveillance of the CNS, by shaping immune responses, not only locally at the meninges, but also at the level of the brain tissue. In this review, we will provide a comprehensive view on our current knowledge about the meningeal lymphatic vasculature, emphasizing its described roles in modulating CNS fluid and macromolecule drainage, meningeal and brain immunity, as well as glial and neuronal function in aging and in AD.
Collapse
Affiliation(s)
- Shanon Rego
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Guadalupe Sanchez
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA.
- Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
27
|
Manukjan N, Majcher D, Leenders P, Caiment F, van Herwijnen M, Smeets HJ, Suidgeest E, van der Weerd L, Vanmierlo T, Jansen JFA, Backes WH, van Oostenbrugge RJ, Staals J, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Hypoxic oligodendrocyte precursor cell-derived VEGFA is associated with blood-brain barrier impairment. Acta Neuropathol Commun 2023; 11:128. [PMID: 37550790 PMCID: PMC10405482 DOI: 10.1186/s40478-023-01627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023] Open
Abstract
Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Daria Majcher
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Hubert J. Smeets
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernst Suidgeest
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
| | - Louise van der Weerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, The Netherlands
| | - Tim Vanmierlo
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jacobus F. A. Jansen
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Walter H. Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Robert J. van Oostenbrugge
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Julie Staals
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
28
|
Iadecola C, Anfray A, Schaeffer S, Hattori Y, Santisteban M, Casey N, Wang G, Strickland M, Zhou P, Holtzman D, Anrather J, Park L. Cell autonomous role of border associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. RESEARCH SQUARE 2023:rs.3.rs-3222611. [PMID: 37577565 PMCID: PMC10418550 DOI: 10.21203/rs.3.rs-3222611/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Apolipoprotein-E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4 we report that border associated macrophages (BAM), myeloid cells closely apposed to neocortical microvessels, are both the source and the target of the ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAM is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Wang
- Feil Family Brain and Mind Research Institute - Weill Cornell Medicine
| | | | | | | | | | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
29
|
Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab 2023; 34:430-445. [PMID: 37357100 PMCID: PMC10365028 DOI: 10.1016/j.tem.2023.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/27/2023]
Abstract
Dysregulation of lipid metabolism has emerged as a central component of many neurodegenerative diseases. Variants of the lipid transport protein, apolipoprotein E (APOE), modulate risk and resilience in several neurodegenerative diseases including late-onset Alzheimer's disease (LOAD). Allelic variants of the gene, APOE, alter the lipid metabolism of cells and tissues and have been broadly associated with several other cellular and systemic phenotypes. Targeting APOE-associated metabolic pathways may offer opportunities to alter disease-related phenotypes and consequently, attenuate disease risk and impart resilience to multiple neurodegenerative diseases. We review the molecular, cellular, and tissue-level alterations to lipid metabolism that arise from different APOE isoforms. These changes in lipid metabolism could help to elucidate disease mechanisms and tune neurodegenerative disease risk and resilience.
Collapse
Affiliation(s)
- Linda G Yang
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zachary M March
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Roxan A Stephenson
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Priyanka S Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.; National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias (CARD), National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Kakae M, Nakajima H, Tobori S, Kawashita A, Miyanohara J, Morishima M, Nagayasu K, Nakagawa T, Shigetomi E, Koizumi S, Mori Y, Kaneko S, Shirakawa H. The astrocytic TRPA1 channel mediates an intrinsic protective response to vascular cognitive impairment via LIF production. SCIENCE ADVANCES 2023; 9:eadh0102. [PMID: 37478173 PMCID: PMC10361588 DOI: 10.1126/sciadv.adh0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 06/20/2023] [Indexed: 07/23/2023]
Abstract
Vascular cognitive impairment (VCI) refers to cognitive alterations caused by vascular disease, which is associated with various types of dementia. Because chronic cerebral hypoperfusion (CCH) induces VCI, we used bilateral common carotid artery stenosis (BCAS) mice as a CCH-induced VCI model. Transient receptor potential ankyrin 1 (TRPA1), the most redox-sensitive TRP channel, is functionally expressed in the brain. Here, we investigated the pathophysiological role of TRPA1 in CCH-induced VCI. During early-stage CCH, cognitive impairment and white matter injury were induced by BCAS in TRPA1-knockout but not wild-type mice. TRPA1 stimulation with cinnamaldehyde ameliorated BCAS-induced outcomes. RNA sequencing analysis revealed that BCAS increased leukemia inhibitory factor (LIF) in astrocytes. Moreover, hydrogen peroxide-treated TRPA1-stimulated primary astrocyte cultures expressed LIF, and culture medium derived from these cells promoted oligodendrocyte precursor cell myelination. Overall, TRPA1 in astrocytes prevents CCH-induced VCI through LIF production. Therefore, TRPA1 stimulation may be a promising therapeutic approach for VCI.
Collapse
Affiliation(s)
- Masashi Kakae
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Department of Clinical Pharmacology and Pharmacotherapy, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Hiroki Nakajima
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Shota Tobori
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Ayaka Kawashita
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Jun Miyanohara
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Misa Morishima
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Pharmacotherapy, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
31
|
Torres-Espin A, Rabadaugh H, Fitzsimons S, Harvey D, Chou A, Lindberg C, Casaletto KB, Goldberger L, Staffaroni AM, Maillard P, Miller BL, DeCarli C, Hinman JD, Ferguson AR, Kramer JH, Elahi FM. Sexually dimorphic differences in angiogenesis markers predict brain aging trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549192. [PMID: 37503183 PMCID: PMC10370093 DOI: 10.1101/2023.07.16.549192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Aberrant angiogenesis could contribute to cognitive impairment, representing a therapeutic target for preventing dementia. However, most angiogenesis studies focus on model organisms. To test the relevance of angiogenesis to human cognitive aging, we evaluated associations of circulating blood markers of angiogenesis with brain aging trajectories in two deeply phenotyped human cohorts (n=435, age 74 + 9) with longitudinal cognitive assessments, biospecimens, structural brain imaging, and clinical data. Machine learning and traditional statistics revealed sex dimorphic associations of plasma angiogenic growth factors with brain aging outcomes. Specifically, angiogenesis is associated with higher executive function and less brain atrophy in younger women (not men), a directionality of association that reverses around age 75. Higher levels of basic fibroblast growth factor, known for pleiotropic effects on multiple cell types, predicted favorable cognitive trajectories. This work demonstrates the relevance of angiogenesis to brain aging with important therapeutic implications for vascular cognitive impairment and dementia.
Collapse
|
32
|
Iadecola C, Smith EE, Anrather J, Gu C, Mishra A, Misra S, Perez-Pinzon MA, Shih AY, Sorond FA, van Veluw SJ, Wellington CL. The Neurovasculome: Key Roles in Brain Health and Cognitive Impairment: A Scientific Statement From the American Heart Association/American Stroke Association. Stroke 2023; 54:e251-e271. [PMID: 37009740 PMCID: PMC10228567 DOI: 10.1161/str.0000000000000431] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
BACKGROUND Preservation of brain health has emerged as a leading public health priority for the aging world population. Advances in neurovascular biology have revealed an intricate relationship among brain cells, meninges, and the hematic and lymphatic vasculature (the neurovasculome) that is highly relevant to the maintenance of cognitive function. In this scientific statement, a multidisciplinary team of experts examines these advances, assesses their relevance to brain health and disease, identifies knowledge gaps, and provides future directions. METHODS Authors with relevant expertise were selected in accordance with the American Heart Association conflict-of-interest management policy. They were assigned topics pertaining to their areas of expertise, reviewed the literature, and summarized the available data. RESULTS The neurovasculome, composed of extracranial, intracranial, and meningeal vessels, as well as lymphatics and associated cells, subserves critical homeostatic functions vital for brain health. These include delivering O2 and nutrients through blood flow and regulating immune trafficking, as well as clearing pathogenic proteins through perivascular spaces and dural lymphatics. Single-cell omics technologies have unveiled an unprecedented molecular heterogeneity in the cellular components of the neurovasculome and have identified novel reciprocal interactions with brain cells. The evidence suggests a previously unappreciated diversity of the pathogenic mechanisms by which disruption of the neurovasculome contributes to cognitive dysfunction in neurovascular and neurodegenerative diseases, providing new opportunities for the prevention, recognition, and treatment of these conditions. CONCLUSIONS These advances shed new light on the symbiotic relationship between the brain and its vessels and promise to provide new diagnostic and therapeutic approaches for brain disorders associated with cognitive dysfunction.
Collapse
|
33
|
Raber J, Silbert LC. Role of white matter hyperintensity in effects of apolipoprotein E on cognitive injury. Front Hum Neurosci 2023; 17:1176690. [PMID: 37275347 PMCID: PMC10237322 DOI: 10.3389/fnhum.2023.1176690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Magnetic Resonance Imaging (MRI) T2-weighted white matter hyperintensity (WMH) is a marker of small vessel cerebrovascular pathology and is of ischemic origin. The prevalence and severity of WMH is associated with cardiovascular risk factors, aging, and cognitive injury in mild cognitive impairment (MCI), vascular dementia, and Alzheimer's disease (AD). WMH especially affects executive function, with additional effects on memory and global cognition. Apolipoprotein E (apoE) plays a role in cholesterol metabolism and neuronal repair after injury. Human and animal studies support a role for apoE in maintaining white matter integrity. In humans, there are three major human apoE isoforms, E2, E3, and E4. Human apoE isoforms differ in risk to develop AD and in association with WMH. In this Mini Review, we propose an increased focus on the role of WMH in cognitive health and cognitive injury and the likely role of apoE and apoE isoform in modulating these effects. We hypothesize that apoE and apoE isoforms play a role in modulating WMH via apoE isoform-dependent effects on oxylipins and 7-ketocholesterol, as well as amyloid related vascular injury, as seen in cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Lisa C. Silbert
- Department of Neurology, Oregon Alzheimer’s Disease Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Veterans Affairs Portland Health Care System, Portland, OR, United States
| |
Collapse
|
34
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
35
|
Uekawa K, Hattori Y, Ahn SJ, Seo J, Casey N, Anfray A, Zhou P, Luo W, Anrather J, Park L, Iadecola C. Border-associated macrophages promote cerebral amyloid angiopathy and cognitive impairment through vascular oxidative stress. RESEARCH SQUARE 2023:rs.3.rs-2719812. [PMID: 37162996 PMCID: PMC10168479 DOI: 10.21203/rs.3.rs-2719812/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Background: Cerebral amyloid angiopathy (CAA) is a devastating condition common in patients with Alzheimer's disease but also observed in the general population. Vascular oxidative stress and neurovascular dysfunction have been implicated in CAA but the cellular source of reactive oxygen species (ROS) and related signaling mechanisms remain unclear. We tested the hypothesis that brain border-associated macrophages (BAM), yolk sac-derived myeloid cells closely apposed to parenchymal and leptomeningeal blood vessels, are the source of radicals through the Aβ-binding innate immunity receptor CD36, leading to neurovascular dysfunction, CAA, and cognitive impairment. Methods: Tg2576 mice and WT littermates were transplanted with CD36 -/- or CD36 +/+ bone marrow at 12-month of age and tested at 15 months. This approach enables the repopulation of perivascular and leptomeningeal compartments with CD36 -/- BAM. Neurovascular function was tested in anesthetized mice equipped with a cranial window in which cerebral blood flow was monitored by laser-Doppler flowmetry. Amyloid pathology and cognitive function were also examined. Results: The increase in blood flow evoked by whisker stimulation (functional hyperemia) or by endothelial and smooth muscle vasoactivity was markedly attenuated in WT®Tg2576 chimeras but was fully restored in CD36 -/- ®Tg2576 chimeras, in which BAM ROS production was suppressed. CAA-associated Aβ 1-40 , but not Aβ 1-42 , was reduced in CD36 -/- ®Tg2576 chimeras. Similarly, CAA, but not parenchymal plaques, was reduced in CD36 -/- ®Tg2576 chimeras. These beneficial vascular effects were associated with cognitive improvement. Finally, CD36 -/- mice were able to more efficiently clear exogenous Aβ 1-40 injected into the neocortex or the striatum. Conclusions: CD36 deletion in BAM suppresses ROS production and rescues the neurovascular dysfunction and damage induced by Aβ. CD36 deletion in BAM also reduced brain Aβ 1-40 and ameliorated CAA without affecting parenchyma plaques. Lack of CD36 enhanced the vascular clearance of exogenous Aβ. Restoration of neurovascular function and attenuation of CAA resulted in a near complete rescue of cognitive function. Collectively, these data implicate CNS BAM in the pathogenesis of CAA and raise the possibility that targeting BAM CD36 is beneficial in CAA and other conditions associated with vascular Aβ deposition and damage.
Collapse
|
36
|
Oue H, Yamazaki Y, Qiao W, Yuanxin C, Ren Y, Kurti A, Shue F, Parsons TM, Perkerson RB, Kawatani K, Wang N, Starling SC, Roy B, Mosneag IE, Aikawa T, Holm ML, Liu CC, Inoue Y, Sullivan PM, Asmann YW, Kim BY, Bu G, Kanekiyo T. LRP1 in vascular mural cells modulates cerebrovascular integrity and function in the presence of APOE4. JCI Insight 2023; 8:e163822. [PMID: 37036005 PMCID: PMC10132158 DOI: 10.1172/jci.insight.163822] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/17/2023] [Indexed: 04/11/2023] Open
Abstract
Cerebrovasculature is critical in maintaining brain homeostasis; its dysregulation often leads to vascular cognitive impairment and dementia (VCID) during aging. VCID is the second most prevalent cause of dementia in the elderly, after Alzheimer's disease (AD), with frequent cooccurrence of VCID and AD. While multiple factors are involved in the pathogenesis of AD and VCID, APOE4 increases the risk for both diseases. A major apolipoprotein E (apoE) receptor, the low-density lipoprotein receptor-related protein 1 (LRP1), is abundantly expressed in vascular mural cells (pericytes and smooth muscle cells). Here, we investigated how deficiency of vascular mural cell LRP1 affects the cerebrovascular system and cognitive performance using vascular mural cell-specific Lrp1-KO mice (smLrp1-/-) in a human APOE3 or APOE4 background. We found that spatial memory was impaired in the 13- to 16-month-old APOE4 smLrp1-/- mice but not in the APOE3 smLrp1-/- mice, compared with their respective littermate control mice. These disruptions in the APOE4 smLrp1-/- mice were accompanied with excess paravascular glial activation and reduced cerebrovascular collagen IV. In addition, blood-brain barrier (BBB) integrity was disrupted in the APOE4 smLrp1-/- mice. Together, our results suggest that vascular mural cell LRP1 modulates cerebrovasculature integrity and function in an APOE genotype-dependent manner.
Collapse
Affiliation(s)
| | | | | | | | - Yingxue Ren
- Department of Quantitative Health Sciences, and
| | | | - Francis Shue
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Tammee M. Parsons
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Ralph B. Perkerson
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | | | | | | | | | | | | | | | - Patrick M. Sullivan
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Betty Y.S. Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Takahisa Kanekiyo
- Department of Neuroscience
- Center for Regenerative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
37
|
Crouzet C, Phan T, Wilson RH, Shin TJ, Choi B. Intrinsic, widefield optical imaging of hemodynamics in rodent models of Alzheimer's disease and neurological injury. NEUROPHOTONICS 2023; 10:020601. [PMID: 37143901 PMCID: PMC10152182 DOI: 10.1117/1.nph.10.2.020601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/30/2023] [Indexed: 05/06/2023]
Abstract
The complex cerebrovascular network is critical to controlling local cerebral blood flow (CBF) and maintaining brain homeostasis. Alzheimer's disease (AD) and neurological injury can result in impaired CBF regulation, blood-brain barrier breakdown, neurovascular dysregulation, and ultimately impaired brain homeostasis. Measuring cortical hemodynamic changes in rodents can help elucidate the complex physiological dynamics that occur in AD and neurological injury. Widefield optical imaging approaches can measure hemodynamic information, such as CBF and oxygenation. These measurements can be performed over fields of view that range from millimeters to centimeters and probe up to the first few millimeters of rodent brain tissue. We discuss the principles and applications of three widefield optical imaging approaches that can measure cerebral hemodynamics: (1) optical intrinsic signal imaging, (2) laser speckle imaging, and (3) spatial frequency domain imaging. Future work in advancing widefield optical imaging approaches and employing multimodal instrumentation can enrich hemodynamic information content and help elucidate cerebrovascular mechanisms that lead to the development of therapeutic agents for AD and neurological injury.
Collapse
Affiliation(s)
- Christian Crouzet
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Thinh Phan
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Biomedical Engineering, Irvine, California, United States
| | - Robert H. Wilson
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Medicine, Irvine, California, United States
| | - Teo Jeon Shin
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- Seoul National University, Department of Pediatric Dentistry and Dental Research Institute, Seoul, Republic of Korea
| | - Bernard Choi
- University of California, Irvine, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
- University of California, Irvine, Department of Biomedical Engineering, Irvine, California, United States
- University of California, Irvine, Department of Surgery, Irvine, California, United States
- University of California, Irvine, Edwards Lifesciences Foundation Cardiovascular Innovation Research Center, California, United States
| |
Collapse
|
38
|
Saavedra J, Nascimento M, Liz MA, Cardoso I. Key brain cell interactions and contributions to the pathogenesis of Alzheimer's disease. Front Cell Dev Biol 2022; 10:1036123. [PMID: 36523504 PMCID: PMC9745159 DOI: 10.3389/fcell.2022.1036123] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/14/2022] [Indexed: 06/22/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide, with the two major hallmarks being the deposition of extracellular β-amyloid (Aβ) plaques and of intracellular neurofibrillary tangles (NFTs). Additionally, early pathological events such as cerebrovascular alterations, a compromised blood-brain barrier (BBB) integrity, neuroinflammation and synaptic dysfunction, culminate in neuron loss and cognitive deficits. AD symptoms reflect a loss of neuronal circuit integrity in the brain; however, neurons do not operate in isolation. An exclusively neurocentric approach is insufficient to understand this disease, and the contribution of other brain cells including astrocytes, microglia, and vascular cells must be integrated in the context. The delicate balance of interactions between these cells, required for healthy brain function, is disrupted during disease. To design successful therapies, it is critical to understand the complex brain cellular connections in AD and the temporal sequence of their disturbance. In this review, we discuss the interactions between different brain cells, from physiological conditions to their pathological reactions in AD, and how this basic knowledge can be crucial for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Joana Saavedra
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Mariana Nascimento
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| | - Márcia A. Liz
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Isabel Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Wang Y, LeDue JM, Murphy TH. Multiscale imaging informs translational mouse modeling of neurological disease. Neuron 2022; 110:3688-3710. [PMID: 36198319 DOI: 10.1016/j.neuron.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Multiscale neurophysiology reveals that simple motor actions are associated with changes in neuronal firing in virtually every brain region studied. Accordingly, the assessment of focal pathology such as stroke or progressive neurodegenerative diseases must also extend widely across brain areas. To derive mechanistic information through imaging, multiple resolution scales and multimodal factors must be included, such as the structure and function of specific neurons and glial cells and the dynamics of specific neurotransmitters. Emerging multiscale methods in preclinical animal studies that span micro- to macroscale examinations fill this gap, allowing a circuit-based understanding of pathophysiological mechanisms. Combined with high-performance computation and open-source data repositories, these emerging multiscale and large field-of-view techniques include live functional ultrasound, multi- and single-photon wide-scale light microscopy, video-based miniscopes, and tissue-penetrating fiber photometry, as well as variants of post-mortem expansion microscopy. We present these technologies and outline use cases and data pipelines to uncover new knowledge within animal models of stroke, Alzheimer's disease, and movement disorders.
Collapse
Affiliation(s)
- Yundi Wang
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Jeffrey M LeDue
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Timothy H Murphy
- University of British Columbia, Department of Psychiatry, Kinsmen Laboratory of Neurological Research, Detwiller Pavilion, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
40
|
Barisano G, Kisler K, Wilkinson B, Nikolakopoulou AM, Sagare AP, Wang Y, Gilliam W, Huuskonen MT, Hung ST, Ichida JK, Gao F, Coba MP, Zlokovic BV. A "multi-omics" analysis of blood-brain barrier and synaptic dysfunction in APOE4 mice. J Exp Med 2022; 219:e20221137. [PMID: 36040482 PMCID: PMC9435921 DOI: 10.1084/jem.20221137] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023] Open
Abstract
Apolipoprotein E4 (APOE4), the main susceptibility gene for Alzheimer's disease, leads to blood-brain barrier (BBB) breakdown in humans and mice. Remarkably, BBB dysfunction predicts cognitive decline and precedes synaptic deficits in APOE4 human carriers. How APOE4 affects BBB and synaptic function at a molecular level, however, remains elusive. Using single-nucleus RNA-sequencing and phosphoproteome and proteome analysis, we show that APOE4 compared with APOE3 leads to an early disruption of the BBB transcriptome in 2-3-mo-old APOE4 knock-in mice, followed by dysregulation in protein signaling networks controlling cell junctions, cytoskeleton, clathrin-mediated transport, and translation in brain endothelium, as well as transcription and RNA splicing suggestive of DNA damage in pericytes. Changes in BBB signaling mechanisms paralleled an early, progressive BBB breakdown and loss of pericytes, which preceded postsynaptic interactome disruption and behavioral deficits that developed 2-5 mo later. Thus, dysregulated signaling mechanisms in endothelium and pericytes in APOE4 mice reflect a molecular signature of a progressive BBB failure preceding changes in synaptic function and behavior.
Collapse
Affiliation(s)
- Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Brent Wilkinson
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Abhay P. Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - William Gilliam
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Mikko T. Huuskonen
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Shu-Ting Hung
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA
| | - Justin K. Ichida
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA
| | - Fan Gao
- Caltech Bioinformatics Resource Center, Caltech, Pasadena, CA
| | - Marcelo P. Coba
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
41
|
Steele OG, Stuart AC, Minkley L, Shaw K, Bonnar O, Anderle S, Penn AC, Rusted J, Serpell L, Hall C, King S. A multi-hit hypothesis for an APOE4-dependent pathophysiological state. Eur J Neurosci 2022; 56:5476-5515. [PMID: 35510513 PMCID: PMC9796338 DOI: 10.1111/ejn.15685] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 01/01/2023]
Abstract
The APOE gene encoding the Apolipoprotein E protein is the single most significant genetic risk factor for late-onset Alzheimer's disease. The APOE4 genotype confers a significantly increased risk relative to the other two common genotypes APOE3 and APOE2. Intriguingly, APOE4 has been associated with neuropathological and cognitive deficits in the absence of Alzheimer's disease-related amyloid or tau pathology. Here, we review the extensive literature surrounding the impact of APOE genotype on central nervous system dysfunction, focussing on preclinical model systems and comparison of APOE3 and APOE4, given the low global prevalence of APOE2. A multi-hit hypothesis is proposed to explain how APOE4 shifts cerebral physiology towards pathophysiology through interconnected hits. These hits include the following: neurodegeneration, neurovascular dysfunction, neuroinflammation, oxidative stress, endosomal trafficking impairments, lipid and cellular metabolism disruption, impaired calcium homeostasis and altered transcriptional regulation. The hits, individually and in combination, leave the APOE4 brain in a vulnerable state where further cumulative insults will exacerbate degeneration and lead to cognitive deficits in the absence of Alzheimer's disease pathology and also a state in which such pathology may more easily take hold. We conclude that current evidence supports an APOE4 multi-hit hypothesis, which contributes to an APOE4 pathophysiological state. We highlight key areas where further study is required to elucidate the complex interplay between these individual mechanisms and downstream consequences, helping to frame the current landscape of existing APOE-centric literature.
Collapse
Affiliation(s)
| | | | - Lucy Minkley
- School of Life SciencesUniversity of SussexBrightonUK
| | - Kira Shaw
- School of Life SciencesUniversity of SussexBrightonUK
| | - Orla Bonnar
- School of Life SciencesUniversity of SussexBrightonUK
| | | | | | | | | | | | - Sarah King
- School of PsychologyUniversity of SussexBrightonUK
| |
Collapse
|
42
|
Cognitive Impairments and blood-brain Barrier Damage in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurochem Res 2022; 47:3817-3828. [DOI: 10.1007/s11064-022-03799-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/07/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
AbstractChronic cerebral hypoperfusion (CCH) is commonly involved in various brain diseases. Tight junction proteins (TJs) are key components constituting the anatomical substrate of the blood-brain barrier (BBB). Changes in cognitive function and BBB after CCH and their relationship need further exploration. To investigate the effect of CCH on cognition and BBB, we developed a bilateral common carotid artery stenosis (BCAS) model in Tie2-GFP mice. Mice manifested cognitive impairments accompanied with increased microglia after the BCAS operation. BCAS mice also exhibited increased BBB permeability at all time points set from D1 to D42. Furthermore, BCAS mice showed reduced expression of TJs 42 d after the operation. In addition, correct entrances of mice in radial arm maze test had a moderate negative correlation with EB extravasation. Our data suggested that BCAS could lead to cognitive deficits, microglia increase and BBB dysfunction characterized by increased BBB permeability and reduced TJs expression level. BBB permeability may be involved in the cognitive impairments induced by CCH.
Collapse
|
43
|
Alexander C, Li T, Hattori Y, Chiu D, Frost GR, Jonas L, Liu C, Anderson CJ, Wong E, Park L, Iadecola C, Li YM. Hypoxia Inducible Factor-1α binds and activates γ-secretase for Aβ production under hypoxia and cerebral hypoperfusion. Mol Psychiatry 2022; 27:4264-4273. [PMID: 35764706 PMCID: PMC9722522 DOI: 10.1038/s41380-022-01676-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023]
Abstract
Hypoxic-ischemic injury has been linked with increased risk for developing Alzheimer's disease (AD). The underlying mechanism of this association is poorly understood. Here, we report distinct roles for hypoxia-inducible factor-1α (Hif-1α) in the regulation of BACE1 and γ-secretase activity, two proteases involved in the production of amyloid-beta (Aβ). We have demonstrated that Hif-1α upregulates both BACE1 and γ-secretase activity for Aβ production in brain hypoxia-induced either by cerebral hypoperfusion or breathing 10% O2. Hif-1α binds to γ-secretase, which elevates the amount of active γ-secretase complex without affecting the level of individual subunits in hypoxic-ischemic mouse brains. Additionally, the expression of full length Hif-1α increases BACE1 and γ-secretase activity in primary neuronal culture, whereas a transcriptionally incompetent Hif-1α variant only activates γ-secretase. These findings indicate that Hif-1α transcriptionally upregulates BACE1 and nontranscriptionally activates γ-secretase for Aβ production in hypoxic-ischemic conditions. Consequently, Hif-1α-mediated Aβ production may be an adaptive response to hypoxic-ischemic injury, subsequently leading to increased risk for AD. Preventing the interaction of Hif-1α with γ-secretase may therefore be a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Courtney Alexander
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Thomas Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Danica Chiu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Georgia R Frost
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Lauren Jonas
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chenge Liu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Corey J Anderson
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Programs of Neurosciences and Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA.
| |
Collapse
|
44
|
Wang X, Jiao B, Liu H, Wang Y, Hao X, Zhu Y, Xu B, Xu H, Zhang S, Jia X, Xu Q, Liao X, Zhou Y, Jiang H, Wang J, Guo J, Yan X, Tang B, Zhao R, Shen L. Machine learning based on Optical Coherence Tomography images as a diagnostic tool for Alzheimer's disease. CNS Neurosci Ther 2022; 28:2206-2217. [PMID: 36089740 PMCID: PMC9627364 DOI: 10.1111/cns.13963] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/03/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023] Open
Abstract
AIMS We mainly evaluate retinal alterations in Alzheimer's disease (AD) patients, investigate the associations between retinal changes with AD biomarkers, and explore an optimal machine learning (ML) model for AD diagnosis based on retinal thickness. METHODS A total of 159 AD patients and 299 healthy controls were enrolled. The retinal parameters of each participant were measured using optical coherence tomography (OCT). Additionally, cognitive impairment severity, brain atrophy, and cerebrospinal fluid (CSF) biomarkers were measured in AD patients. RESULTS AD patients demonstrated a significant decrease in the average, superior, and inferior quadrant peripapillary retinal nerve fiber layer, macular retinal nerve fiber layer, ganglion cell layer (GCL), inner plexiform layer (IPL) thicknesses, as well as total macular volume (TMV) (all p < 0.05). Moreover, TMV was positively associated with Mini-Mental State Examination and Montreal Cognitive Assessment scores, IPL thickness was correlated negatively with the medial temporal lobe atrophy score, and the GCL thickness was positively correlated with CSF Aβ42 /Aβ40 and negatively associated with p-tau level. Based on the significantly decreased OCT variables between both groups, the XGBoost algorithm exhibited the best diagnostic performance for AD, whose four references, including accuracy, area under the curve, f1 score, and recall, ranged from 0.69 to 0.74. Moreover, the macular retinal thickness exhibited an absolute superiority for AD diagnosis compared with other enrolled variables in all ML models. CONCLUSION We identified the retinal alterations in AD patients and found that macular thickness and volume were associated with AD severity and biomarkers. Furthermore, we confirmed that OCT combined with ML could serve as a potential diagnostic tool for AD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Bin Jiao
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Hui Liu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yaqin Wang
- Health Management Center, the Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoli Hao
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yuan Zhu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Bei Xu
- Eye Center of Xiangya HospitalCentral South UniversityChangshaChina
| | - Huizhuo Xu
- Eye Center of Xiangya HospitalCentral South UniversityChangshaChina
| | - Sizhe Zhang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina
| | - Xiaoliang Jia
- School of Computer Science and EngineeringCentral South UniversityChangshaChina
| | - Qian Xu
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Xinxin Liao
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina,Department of Geriatrics, Xiangya HospitalCentral South UniversityChangshaChina
| | - Yafang Zhou
- National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina,Department of Geriatrics, Xiangya HospitalCentral South UniversityChangshaChina
| | - Hong Jiang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Junling Wang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Jifeng Guo
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Xinxiang Yan
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Beisha Tang
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina
| | - Rongchang Zhao
- School of Computer Science and EngineeringCentral South UniversityChangshaChina
| | - Lu Shen
- Department of Neurology, Xiangya HospitalCentral South UniversityChangshaChina,National Clinical Research Center for Geriatric DisordersCentral South UniversityChangshaChina,Engineering Research Center of Hunan Province in Cognitive Impairment DisordersCentral South UniversityChangshaChina,Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic DiseasesChangshaChina,Key Laboratory of Hunan Province in Neurodegenerative DisordersCentral South UniversityChangshaChina,Key Laboratory of Organ InjuryAging and Regenerative Medicine of Hunan ProvinceChangshaChina
| |
Collapse
|
45
|
Alfieri A, Koudelka J, Li M, Scheffer S, Duncombe J, Caporali A, Kalaria RN, Smith C, Shah AM, Horsburgh K. Nox2 underpins microvascular inflammation and vascular contributions to cognitive decline. J Cereb Blood Flow Metab 2022; 42:1176-1191. [PMID: 35102790 PMCID: PMC9207496 DOI: 10.1177/0271678x221077766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022]
Abstract
Chronic microvascular inflammation and oxidative stress are inter-related mechanisms underpinning white matter disease and vascular cognitive impairment (VCI). A proposed mediator is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2), a major source of reactive oxygen species (ROS) in the brain. To assess the role of Nox2 in VCI, we studied a tractable model with white matter pathology and cognitive impairment induced by bilateral carotid artery stenosis (BCAS). Mice with genetic deletion of Nox2 (Nox2 KO) were compared to wild-type (WT) following BCAS. Sustained BCAS over 12 weeks in WT mice induced Nox2 expression, indices of microvascular inflammation and oxidative damage, along with white matter pathology culminating in a marked cognitive impairment, which were all protected by Nox2 genetic deletion. Neurovascular coupling was impaired in WT mice post-BCAS and restored in Nox2 KO mice. Increased vascular expression of chemoattractant mediators, cell-adhesion molecules and endothelial activation factors in WT mice post-BCAS were ameliorated by Nox2 deficiency. The clinical relevance was confirmed by increased vascular Nox2 and indices of microvascular inflammation in human post-mortem subjects with cerebral vascular disease. Our results support Nox2 activity as a critical determinant of VCI, whose targeting may be of therapeutic benefit in cerebral vascular disease.
Collapse
Affiliation(s)
- Alessio Alfieri
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- National Heart and Lung Institute, Vascular Science, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, UK
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sanny Scheffer
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Andrea Caporali
- British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rajesh N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle-Upon-Tyne, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
46
|
Zhang Y, Zhao B, Lai Q, Li Q, Tang X, Zhang Y, Pan Z, Gao Q, Zhong Z. Chronic cerebral hypoperfusion and blood-brain barrier disruption in uninjured brain areas of rhesus monkeys subjected to transient ischemic stroke. J Cereb Blood Flow Metab 2022; 42:1335-1346. [PMID: 35137610 PMCID: PMC9207497 DOI: 10.1177/0271678x221078065] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Blood-brain barrier (BBB) disruption is a pivotal pathophysiological process in ischemic stroke. Although temporal changes in BBB permeability during the acute phase have been widely studied, little is known about the chronic phase of cerebrovascular changes that may have a large impact on the long-term outcome. Therefore, this study was aimed to measure cerebral vascular abnormalities using CT perfusion in nine rhesus monkeys subjected to transient middle cerebral artery occlusion (tMCAO) for ≥1 year (MCAO-1Y+). The level of cerebral perfusion demonstrated by mean transit time was significantly higher in the ipsilateral caudate nucleus, white matter, thalamus, hippocampus, and contralateral thalamus in MCAO-1Y+ compared with the other nine age-matched control monkeys. The increase in BBB permeability measured through the permeability surface was found in the same ten regions of interest ipsilaterally and contralaterally. We also found decreased levels of Aβ 42/40 ratio in the cerebrospinal fluid (CSF), suggesting a potential link between post-MCAO cognitive decline and Aβ metabolism. Overall, we demonstrated significant cerebral hypoperfusion, BBB disruption, and CSF Aβ decrease during the rehabilitation stage of ischemic stroke in a non-human primate model. Future studies are needed to elucidate the cause-effect relationship between cerebrovascular disruptions and long-term neurological deficits.
Collapse
Affiliation(s)
- Yingqian Zhang
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Bangcheng Zhao
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Lai
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institute, Chengdu, China
| | - Qinxi Li
- School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Xun Tang
- Sichuan SAFE Pharmaceutical Technology Company Limited, Chengdu, China
| | - Yinbing Zhang
- Sichuan SAFE Pharmaceutical Technology Company Limited, Chengdu, China
| | - Zhixiang Pan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Gao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihui Zhong
- Laboratory of Nonhuman Primate Disease Modeling Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
47
|
Wang YL, Sun M, Wang FZ, Wang X, Jia Z, Zhang Y, Li R, Jiang J, Wang L, Li W, Sun Y, Chen J, Zhang C, Shi B, Liu J, Liu X, Xu J. Mediation of the APOE Associations With Cognition Through Cerebral Blood Flow: The CIBL Study. Front Aging Neurosci 2022; 14:928925. [PMID: 35847686 PMCID: PMC9279129 DOI: 10.3389/fnagi.2022.928925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
Background The ε4 allele of the apolipoprotein E (APOE) gene is a strong genetic risk factor for aging-related cognitive decline. However, the causal connection between ε4 alleles and cognition is not well understood. The objective of this study was to identify the roles of cerebral blood flow (CBF) in cognitive-related brain areas in mediating the associations of APOE with cognition. Methods The multiple linear regression analyses were conducted on 369 subjects (mean age of 68.8 years; 62.9% of women; 29.3% of APOE ε4 allele carriers). Causal mediation analyses with 5,000 bootstrapped iterations were conducted to explore the mediation effects. Result APOE ε4 allele was negatively associated with cognition (P < 0.05) and CBF in the amygdala, hippocampus, middle temporal gyrus, posterior cingulate, and precuneus (all P < 0.05). The effect of the APOE genotype on cognition was partly mediated by the above CBF (all P < 0.05). Conclusion CBF partially mediates the potential links between APOE genotype and cognition. Overall, the APOE ε4 allele may lead to a dysregulation of the vascular structure and function with reduced cerebral perfusion, which in turn leads to cognitive impairment.
Collapse
Affiliation(s)
- Yan-Li Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fang-Ze Wang
- Department of Cardiology, Weifang People’s Hospital, Weifang Medical University, Weifang, China
| | - Xiaohong Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Ziyan Jia
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Runzhi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Linlin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yongan Sun
- Department of Neurology, Peking University First Hospital, Peking University, Beijing, China
| | - Jinglong Chen
- Division of Neurology, Department of Geriatrics, National Clinical Key Specialty, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Cuicui Zhang
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Baolin Shi
- Department of Neurology, Weifang People’s Hospital, Weifang Medical University, Weifang, China
| | - Jianjian Liu
- Department of Neurology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Xiangrong Liu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Jun Xu,
| |
Collapse
|
48
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
49
|
Zhou C, Sun P, Xu Y, Chen Y, Huang Y, Hamblin MH, Foley L, Hitchens TK, Li S, Yin K. Genetic Deficiency of MicroRNA-15a/16-1 Confers Resistance to Neuropathological Damage and Cognitive Dysfunction in Experimental Vascular Cognitive Impairment and Dementia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104986. [PMID: 35403823 PMCID: PMC9189640 DOI: 10.1002/advs.202104986] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/22/2022] [Indexed: 05/24/2023]
Abstract
Chronic cerebral hypoperfusion-derived brain damage contributes to the progression of vascular cognitive impairment and dementia (VCID). Cumulative evidence has shown that microRNAs (miRs) are emerging as novel therapeutic targets for CNS disorders. In this study, it is sought to determine the regulatory role of miR-15a/16-1 in VCID. It is found that miR-15a/16-1 knockout (KO) mice exhibit less cognitive and sensorimotor deficits following VCID. Genetic deficiency of miR-15a/16-1 in VCID mice also mitigate myelin degeneration, axonal injury, and neuronal loss. Mechanistically, miR-15a/16-1 binds to the 3'-UTR of AKT3 and IL-10RA. Genetic deletion of miR-15a/16-1 increases AKT3 and IL-10RA expression in VCID brains, and intranasal delivery of AKT3 and IL-10RA siRNA-loaded nanoparticles partially reduce brain protection and cognitive recovery in miR-15a/16-1 KO mice after VCID. In conclusion, the miR-15a/16-1-IL/10RA/AKT3 axis plays a critical role in regulating vascular brain damage and cognitive decline after VCID. Targeting miR-15a/16-1 is a novel therapeutic approach for the treatment of VCID.
Collapse
Affiliation(s)
- Chao Zhou
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Ping Sun
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
| | - Yuang Chen
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Yixian Huang
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Milton H. Hamblin
- Tulane University Health Sciences CenterTulane UniversityNew OrleansLA70112USA
| | - Lesley Foley
- Animal Imaging CenterDepartment of NeurobiologyUniversity of Pittsburgh School of MedicinePittsburghPA15203USA
| | - T. Kevin Hitchens
- Animal Imaging CenterDepartment of NeurobiologyUniversity of Pittsburgh School of MedicinePittsburghPA15203USA
| | - Song Li
- Center for PharmacogeneticsUniversity of Pittsburgh School of PharmacyPittsburghPA15213USA
| | - Ke‐Jie Yin
- Pittsburgh Institute of Brain Disorders & RecoveryDepartment of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPA15213USA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPA15240USA
| |
Collapse
|
50
|
Khoury MA, Bahsoun MA, Fadhel A, Shunbuli S, Venkatesh S, Ghazvanchahi A, Mitha S, Chan K, Fornazzari LR, Churchill NW, Ismail Z, Munoz DG, Schweizer TA, Moody AR, Fischer CE, Khademi A. Delusional Severity Is Associated with Abnormal Texture in FLAIR MRI. Brain Sci 2022; 12:600. [PMID: 35624987 PMCID: PMC9139341 DOI: 10.3390/brainsci12050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
Background: This study examines the relationship between delusional severity in cognitively impaired adults with automatically computed volume and texture biomarkers from the Normal Appearing Brain Matter (NABM) in FLAIR MRI. Methods: Patients with mild cognitive impairment (MCI, n = 24) and Alzheimer’s Disease (AD, n = 18) with delusions of varying severities based on Neuropsychiatric Inventory-Questionnaire (NPI-Q) (1—mild, 2—moderate, 3—severe) from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) were analyzed for this task. The NABM region, which is gray matter (GM) and white matter (WM) combined, was automatically segmented in FLAIR MRI volumes with intensity standardization and thresholding. Three imaging biomarkers were computed from this region, including NABM volume and two texture markers called “Integrity” and “Damage”. Together, these imaging biomarkers quantify structural changes in brain volume, microstructural integrity and tissue damage. Multivariable regression was used to investigate relationships between imaging biomarkers and delusional severities (1, 2 and 3). Sex, age, education, APOE4 and baseline cerebrospinal fluid (CSF) tau were included as co-variates. Results: Biomarkers were extracted from a total of 42 participants with longitudinal time points representing 164 imaging volumes. Significant associations were found for all three NABM biomarkers between delusion level 3 and level 1. Integrity was also sensitive enough to show differences between delusion level 1 and delusion level 2. A significant specified interaction was noted with severe delusions (level 3) and CSF tau for all imaging biomarkers (p < 0.01). APOE4 homozygotes were also significantly related to the biomarkers. Conclusion: Cognitively impaired older adults with more severe delusions have greater global brain disease burden in the WM and GM combined (NABM) as measured using FLAIR MRI. Relative to patients with mild delusions, tissue degeneration in the NABM was more pronounced in subjects with higher delusional symptoms, with a significant association with CSF tau. Future studies are required to establish potential tau-associated mechanisms of increased delusional severity.
Collapse
Affiliation(s)
- Marc A. Khoury
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
| | - Mohamad-Ali Bahsoun
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Electrical, Computer and Biomedical Engineering Department, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Ayad Fadhel
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
| | - Shukrullah Shunbuli
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
| | - Saanika Venkatesh
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| | - Abdollah Ghazvanchahi
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Electrical, Computer and Biomedical Engineering Department, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Samir Mitha
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Electrical, Computer and Biomedical Engineering Department, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Karissa Chan
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Electrical, Computer and Biomedical Engineering Department, Ryerson University, Toronto, ON M5B 2K3, Canada
| | - Luis R. Fornazzari
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Division of Neurology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Nathan W. Churchill
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zahinoor Ismail
- Departments of Psychiatry, Clinical Neurosciences, and Community Health Sciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - David G. Munoz
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tom A. Schweizer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Alan R. Moody
- Department of Medical Imaging, University of Toronto, Toronto, ON M5T 1W7, Canada;
| | - Corinne E. Fischer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - April Khademi
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON M5V 1T8, Canada; (M.A.K.); (A.F.); (S.S.); (S.V.); (L.R.F.); (N.W.C.); (D.G.M.); (T.A.S.); (A.K.)
- Institute for Biomedical Engineering, Science & Tech (iBEST), a Partnership between St. Michael’s Hospital and Ryerson University, Toronto, ON M5V 1T8, Canada; (M.-A.B.); (A.G.); (S.M.); (K.C.)
- Electrical, Computer and Biomedical Engineering Department, Ryerson University, Toronto, ON M5B 2K3, Canada
| |
Collapse
|