1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Mondal R, Deb S, Shome G, Sarkar V, Lahiri D, Datta SS, Benito-León J. Molecular dynamics of amyloid-β transport in Alzheimer's disease: Exploring therapeutic plasma exchange with albumin replacement - Current insights and future perspectives. Neurologia 2025; 40:306-328. [PMID: 40280630 DOI: 10.1016/j.nrleng.2025.03.008] [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: 05/04/2023] [Accepted: 11/07/2023] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The complex process of amyloid-β (Aβ) transportation across the blood-brain and blood-cerebrospinal fluid barriers is crucial for preventing Aβ accumulation, which linked to dementia and neurodegeneration. This review explores therapeutic plasma exchange with albumin replacement in Alzheimer's disease, based on the dynamics of amyloid-β between the brain, plasma, and cerebrospinal fluid. METHODOLOGY A comprehensive literature review was conducted using PubMed/Medline, Cochrane Library, and open databases (bioRxiv, MedRixv, preprint.org) up to April 30, 2023. The first search utilized the following MeSH terms and keywords: 'Plasma Exchange', 'Plasmapheresis', 'Therapeutic plasma exchange', 'Apheresis', 'Aβ', 'p-tau', 'Total-tau', 'Alzheimer's disease', 'Cognitive dysfunction', 'neurodegenerative diseases', 'centrifugation', 'membranous', and 'filtration' in the Title/Abstract, yielding 146 results. A second search with the keywords: 'Albumin', 'Aβ', 'BBB', 'Alzheimer's dementia', and 'Nerve degeneration' resulted in 125 additional articles for analysis. Finally, a third search using keywords: 'Albumin structural domains', 'Albumin-Aβ interactions', 'Albumin-endothelial interactions', and 'Post-Translational Modification' produced 193 results for further review. RESULTS/DISCUSSION Therapeutic plasma exchange shows potential as a disease-modifying therapy for dementia, specifically for Alzheimer's disease. Additionally, the promising role of albumin supplementation in cognitive improvement has attracted attention. However, clinical evidence supporting therapeutic plasma exchange for dementia remains limited, necessitating further research and development to mitigate potential adverse effects. A deeper understanding of the molecular dynamics of Aβ transportation and the mechanisms of therapeutic plasma exchange is essential. A critical evaluation of existing evidence highlights the importance of balancing potential benefits with associated risks, which will guide the development and application of these treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- R Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - S Deb
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - G Shome
- Department of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - V Sarkar
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - D Lahiri
- Baycrest Academy of Research and Education, Toronto, Canada; Rotman Research Institute, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Canada; Department of Neurology, Institute of Neurosciences, Kolkata, India
| | - S S Datta
- Department of Transfusion Medicine, Tata Medical Center, Kolkata 700160, India
| | - J Benito-León
- Department of Neurology, University Hospital "12 de Octubre", Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
3
|
Rozeboom A, Broekaart DWM, Anink JJ, Boonkamp L, Idema S, Teunissen CE, Aronica E, Gorter JA, van Vliet EA. Cellular expression of low-density lipoprotein receptor-related protein 1 and amyloid beta deposition in human and rat epileptogenic brain. Exp Neurol 2025; 386:115149. [PMID: 39842492 DOI: 10.1016/j.expneurol.2025.115149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Decreased capillary expression of low-density lipoprotein receptor-related protein 1 (LRP1) has been linked to increased brain amyloid beta (Aβ) accumulation in Alzheimer's disease (AD). Aβ accumulation has also been observed in (a subset of) temporal lobe epilepsy (TLE) patients, suggesting a potential link between epilepsy and AD. This study examines cellular LRP1 expression in human and rat epileptogenic brain tissue to explore LRP1's role in epilepsy. LRP1 expression and localization were analyzed in hippocampal sections from patients with status epilepticus (SE, n = 12), TLE (n = 12), autopsy controls (n = 20), and AD (n = 10) using immunohistochemistry. Soluble Aβ levels and deposits were compared across TLE, AD, and control tissues. LRP1 expression was also studied in an electrical post-SE rat model of TLE. Decreased capillary LRP1 expression was found in both human and rat brain tissue (SE and TLE). Higher LRP1 expression was detected in CA1 neurons (only in human TLE) and glial cells (SE and TLE). Aβ deposits were observed in only one out of 12 TLE patients, and soluble Aβ levels were not significantly elevated. In contrast, AD patients showed decreased capillary LRP1 expression accompanied by Aβ plaques and increased soluble Aβ40/42 levels. The significant reduction in LRP1 expression in brain capillaries in both adult human and rat TLE was not clearly associated with notable Aβ accumulation implying that alternative amyloid clearance mechanisms beyond LRP1 in blood vessels might be at play. It also supports previous findings indicating that Aβ pathology may be less prominent in adult TLE than some studies suggest.
Collapse
Affiliation(s)
- Annemieke Rozeboom
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Diede W M Broekaart
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lynn Boonkamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Sander Idema
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Erwin A van Vliet
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
4
|
Wada Y, Inoko M, Ishihara K, Fukumoto K, Tsurudome Y, Horiguchi M, Fujimura A, Ushijima K. Aging Reduces ATP-Binding Cassette Transporter Expression in Brain Microvessels of Mice. Pharmaceuticals (Basel) 2025; 18:191. [PMID: 40006002 PMCID: PMC11859312 DOI: 10.3390/ph18020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Background: ATP-binding cassette (ABC) transporters are expressed in the vascular walls of brain capillaries and remove toxic chemicals from the brain. The expression of ABC transporters in peripheral organs is transcriptionally regulated by clock genes and exhibits 24 h periodic fluctuations. In addition, clock gene outputs diminish with aging. In this study, we evaluated whether the expression of ABC transporters in the blood-brain barrier (BBB) of young mice had a 24 h cycle, and whether the expression of ABC transporters in the BBB decreased with age. Methods: Brain microvascular (BMV) fractions from the cerebral cortex of male C57BL/6J mice were prepared using dextran. BMV fractions from young mice (12 weeks old) were prepared every four hours to evaluate 24 h rhythmicity. BMV fractions from both young and aged mice (85 weeks old) were prepared when protein expression peaked (Zeitgeber Time 5). Protein and mRNA expression of ABC transporters in BMV fractions were measured. Results: In young mice, protein expression of P-glycoprotein, breast cancer resistance protein, and multidrug resistance protein 4 showed time-dependent variations with a peak in the light phase (Zeitgeber Time 5); mRNA expression showed no time-dependent variation. The protein expression of these transporters was lower in the BBB of aged mice than in that of young mice, although mRNA expression did not differ between young and aged mice. Conclusions: ABC transporter protein expression levels in BMV endothelial cells decreased with aging; however, mRNA levels did not change, which suggests changes in protein expression did not result from diminished clock gene output. Further studies are needed to elucidate the mechanisms by which ABC transporter expression in the BBB decreases with aging.
Collapse
Affiliation(s)
- Yukiyo Wada
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Masaki Inoko
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Kanako Ishihara
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Karin Fukumoto
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Yuya Tsurudome
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
| | - Michiko Horiguchi
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Department of Pharmaceutical Engineering, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan
| | - Akio Fujimura
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Division of Clinical Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Kentaro Ushijima
- Division of Pharmaceutics, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Yamaguchi 756-0884, Japan (Y.T.); (M.H.)
- Division of Clinical Pharmacology, Jichi Medical University, Tochigi 329-0498, Japan
| |
Collapse
|
5
|
Nogueira Pinto H, Zarekiani P, de Vries HE. Neuroglia and the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:127-141. [PMID: 40122621 DOI: 10.1016/b978-0-443-19104-6.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The blood-brain barrier (BBB) is a highly dynamic and complex structure, present throughout the brain vasculature, that safeguards the brain against blood-borne insults. Neuroglial cells play a major role in its development, function, and homeostasis of the BBB by establishing intricate interactions via direct cell-cell contacts and paracrine signaling. Astrocytes, pericytes, oligodendrocytes, and microglia, alongside specialized brain endothelial cells, orchestrate key events in the brain in health and disease, which can be partially recapitulated by in vitro and in vivo models for biomedical research. This chapter presents a detailed description of the main cellular and molecular mechanisms that govern the neuroglia-BBB crosstalk and the available models for its investigation, emphasizing the importance of each cell population and the synergistic roles they play in the brain.
Collapse
Affiliation(s)
- Henrique Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands
| | - Parand Zarekiani
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Helga E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; MS Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Testa G, Giannelli S, Staurenghi E, Cecci R, Floro L, Gamba P, Sottero B, Leonarduzzi G. The Emerging Role of PCSK9 in the Pathogenesis of Alzheimer's Disease: A Possible Target for the Disease Treatment. Int J Mol Sci 2024; 25:13637. [PMID: 39769398 PMCID: PMC11727734 DOI: 10.3390/ijms252413637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease mainly caused by β-amyloid (Aβ) accumulation in the brain. Among the several factors that may concur to AD development, elevated cholesterol levels and brain cholesterol dyshomeostasis have been recognized to play a relevant role. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a protein primarily known to regulate plasma low-density lipoproteins (LDLs) rich in cholesterol and to be one of the main causes of familial hypercholesterolemia. In addition to that, PCSK9 is also recognized to carry out diverse important activities in the brain, including control of neuronal differentiation, apoptosis, and, importantly, LDL receptors functionality. Moreover, PCSK9 appeared to be directly involved in some of the principal processes responsible for AD development, such as inflammation, oxidative stress, and Aβ deposition. On these bases, PCSK9 management might represent a promising approach for AD treatment. The purpose of this review is to elucidate the role of PCSK9, whether or not cholesterol-related, in AD pathogenesis and to give an updated overview of the most innovative therapeutic strategies developed so far to counteract the pleiotropic activities of both humoral and brain PCSK9, focusing in particular on their potentiality for AD management.
Collapse
Affiliation(s)
- Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
- Division of Neurology Vand Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Rebecca Cecci
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Lucrezia Floro
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, 10043 Orbassano, Italy; (G.T.); (S.G.); (E.S.); (R.C.); (L.F.); (P.G.); (G.L.)
| |
Collapse
|
7
|
Cyr B, Curiel Cid R, Loewenstein D, Vontell RT, Dietrich WD, Keane RW, de Rivero Vaccari JP. The Inflammasome Adaptor Protein ASC in Plasma as a Biomarker of Early Cognitive Changes. Int J Mol Sci 2024; 25:7758. [PMID: 39063000 PMCID: PMC11276719 DOI: 10.3390/ijms25147758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a group of symptoms including memory loss, language difficulties, and other types of cognitive and functional impairments that affects 57 million people worldwide, with the incidence expected to double by 2040. Therefore, there is an unmet need to develop reliable biomarkers to diagnose early brain impairments so that emerging interventions can be applied before brain degeneration. Here, we performed biomarker analyses for apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), and amyloid-β 42/40 (Aβ42/40) ratio in the plasma of older adults. Participants had blood drawn at baseline and underwent two annual clinical and cognitive evaluations. The groups tested either cognitively normal on both evaluations (NN), cognitively normal year 1 but cognitively impaired year 2 (NI), or cognitively impaired on both evaluations (II). ASC was elevated in the plasma of the NI group compared to the NN and II groups. Additionally, Aβ42 was increased in the plasma in the NI and II groups compared to the NN group. Importantly, the area under the curve (AUC) for ASC in participants older than 70 years old in NN vs. NI groups was 0.81, indicating that ASC is a promising plasma biomarker for early detection of cognitive decline.
Collapse
Affiliation(s)
- Brianna Cyr
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Rosie Curiel Cid
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | - David Loewenstein
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
| | | | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
| | - Robert W. Keane
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA; (B.C.); (W.D.D.); (R.W.K.)
- Center for Cognitive Neuroscience and Aging, University of Miami, Miami, FL 33136, USA; (R.C.C.); (D.L.)
- Department of Physiology and Biophysics, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
8
|
Munsterman D, Falcione S, Long R, Boghozian R, Joy T, Camicioli R, Smith EE, Jickling GC. Cerebral amyloid angiopathy and the immune system. Alzheimers Dement 2024; 20:4999-5008. [PMID: 38881491 PMCID: PMC11247707 DOI: 10.1002/alz.13826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 06/18/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of amyloid protein in the walls of cerebral blood vessels. This deposition of amyloid causes damage to the cerebral vasculature, resulting in blood-brain barrier disruption, cerebral hemorrhage, cognitive decline, and dementia. The role of the immune system in CAA is complex and not fully understood. While the immune system has a clear role in the rare inflammatory variants of CAA (CAA related inflammation and Abeta related angiitis), the more common variants of CAA also have immune system involvement. In a protective role, immune cells may facilitate the clearance of beta-amyloid from the cerebral vasculature. The immune system can also contribute to CAA pathology, promoting vascular injury, blood-brain barrier breakdown, inflammation, and progression of CAA. In this review, we summarize the role of the immune system in CAA, including the potential of immune based treatment strategies to slow vascular disease in CAA and associated cognitive impairment, white matter disease progression, and reduce the risk of cerebral hemorrhage. HIGHLIGHTS: The immune system has a role in cerebral amyloid angiopathy (CAA) which is summarized in this review. There is an inflammatory response to beta-amyloid that may contribute to brain injury and cognitive impairment. Immune cells may facilitate the clearance of beta-amyloid from the cerebral vasculature. Improved understanding of the immune system in CAA may afford novel treatment to improve outcomes in patients with CAA.
Collapse
Affiliation(s)
| | - Sarina Falcione
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | - Rebecca Long
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Twinkle Joy
- Division of NeurologyUniversity of AlbertaEdmontonAlbertaCanada
| | | | - Eric E. Smith
- Clinical NeurosciencesHotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | | |
Collapse
|
9
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
10
|
Badaut J, Ghersi-Egea JF, Thorne RG, Konsman JP. Blood-brain borders: a proposal to address limitations of historical blood-brain barrier terminology. Fluids Barriers CNS 2024; 21:3. [PMID: 38183042 PMCID: PMC10770911 DOI: 10.1186/s12987-023-00478-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/11/2023] [Indexed: 01/07/2024] Open
Abstract
Many neuroscientists use the term Blood-Brain Barrier (BBB) to emphasize restrictiveness, often equating or reducing the notion of BBB properties to tight junction molecules physically sealing cerebral endothelial cells, rather than pointing out the complexity of this biological interface with respect to its selectivity and variety of exchange between the general blood circulation and the central nervous tissue. Several authors in the field find it unfortunate that the exquisitely dynamic interfaces between blood and brain continue to be viewed primarily as obstructive barriers to transport. Although the term blood-brain interface is an excellent descriptor that does not convey the idea of a barrier, it is important and preferable for the spreading of an idea beyond specialist communities to try to appeal to well-chosen metaphors. Recent evidence reviewed here indicates that blood-brain interfaces are more than selective semi-permeable membranes in that they display many dynamic processes and complex mechanisms for communication. They are thus more like 'geopolitical borders'. Furthermore, some authors working on blood-brain interface-relevant issues have started to use the word border, for example in border-associated macrophages. Therefore, we suggest adopting the term Blood-Brain Border to better communicate the flexibility of and movement across blood-brain interfaces.
Collapse
Affiliation(s)
- Jerome Badaut
- Brain Molecular Imaging Lab, UMR 5536, CNRS, RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
- Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Jean-François Ghersi-Egea
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR 5292, Lyon-1 University, Bron, France.
| | - Robert G Thorne
- Denali Therapeutics, Inc, 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
- Department of Pharmaceutics, University of Minnesota, 9-177 Weaver-Densford Hall, 308 Harvard St. SE, Minneapolis, MN, 55455, USA.
| | - Jan Pieter Konsman
- UMR 5164, CNRS, ImmunoConcEpT, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
| |
Collapse
|
11
|
Xiao X, Jiang H, Wei H, Zhou Y, Ji X, Zhou C. Endothelial Senescence in Neurological Diseases. Aging Dis 2023; 14:2153-2166. [PMID: 37199574 PMCID: PMC10676791 DOI: 10.14336/ad.2023.0226-1] [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: 12/15/2022] [Accepted: 02/26/2023] [Indexed: 05/19/2023] Open
Abstract
Endothelial cells, which are highly dynamic cells essential to the vascular network, play an indispensable role in maintaining the normal function of the body. Several lines of evidence indicate that the phenotype associated with senescent endothelial cells causes or promotes some neurological disorders. In this review, we first discuss the phenotypic changes associated with endothelial cell senescence; subsequently, we provide an overview of the molecular mechanisms of endothelial cell senescence and its relationship with neurological disorders. For refractory neurological diseases such as stroke and atherosclerosis, we intend to provide some valid clues and new directions for clinical treatment options.
Collapse
Affiliation(s)
- Xuechun Xiao
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Huimin Wei
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Sun L, Wang L, Ye KX, Wang S, Zhang R, Juan Z, Feng L, Min S. Endothelial Glycocalyx in Aging and Age-related Diseases. Aging Dis 2023; 14:1606-1617. [PMID: 37196119 PMCID: PMC10529737 DOI: 10.14336/ad.2023.0131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 05/19/2023] Open
Abstract
The worldwide population is aging exponentially, creating burdens to patients, their families and society. Increasing age is associated with higher risk of a wide range of chronic diseases, and aging of the vascular system is closely linked to the development of many age-related diseases. Endothelial glycocalyx is a layer of proteoglycan polymers on the surface of the inner lumen of blood vessels. It plays an important role in maintaining vascular homeostasis and protecting various organ functions. Endothelial glycocalyx loss happens through the aging process and repairing the endothelial glycocalyx may alleviate the symptoms of age-related diseases. Given the important role of the glycocalyx and its regenerative properties, it is posited that the endothelial glycocalyx may be a potential therapeutic target for aging and age-related diseases and repairing endothelial glycocalyx could play a role in the promotion of healthy aging and longevity. Here, we review the composition, function, shedding, and manifestation of the endothelial glycocalyx in aging and age-related diseases, as well as regeneration of endothelial glycocalyx.
Collapse
Affiliation(s)
- Lina Sun
- School of Anesthesiology, Weifang Medical University, Weifang, China.
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Lingyan Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Kaisy Xinhong Ye
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Shoushi Wang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Rui Zhang
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Zhaodong Juan
- School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Lei Feng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
13
|
Liu LL, Shen X, Gu H, Zhao G, Du Y, Zheng W. High affinity of β-amyloid proteins to cerebral capillaries: implications in chronic lead exposure-induced neurotoxicity in rats. Fluids Barriers CNS 2023; 20:32. [PMID: 37122007 PMCID: PMC10150519 DOI: 10.1186/s12987-023-00432-5] [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: 12/09/2022] [Accepted: 04/10/2023] [Indexed: 05/02/2023] Open
Abstract
Lead (Pb) is a known environmental risk factor in the etiology of Alzheimer's disease (AD). The existing reports suggest that Pb exposure increases beta-amyloid (Aβ) levels in brain tissues and cerebrospinal fluid (CSF) and facilitates the formation of amyloid plaques, which is a pathological hallmark for AD. Pb exposure has long been associated with cerebral vasculature injury. Yet it remained unclear if Pb exposure caused excessive Ab buildup in cerebral vasculature, which may damage the blood-brain barrier and cause abnormal Ab accumulation. This study was designed to investigate the impact of chronic Pb exposure on Aβ accumulation in cerebral capillary and the expression of low-density lipoprotein receptor protein-1 (LRP1), a critical Aβ transporter, in brain capillary and parenchyma. Sprague-Dawley rats received daily oral gavage at doses of 0, 14 (low-dose), and 27 (high-dose) mg Pb/kg as Pb acetate, 5 d/wk, for 4 or 8 wks. At the end of Pb exposure, a solution containing Aβ40 was infused into the brain via the cannulated internal carotid artery. Data by ELISA showed a strikingly high affinity of Ab to cerebral vasculature, which was approximately 7-14 times higher than that to the parenchymal fractions collected from control brains. Pb exposure further aggravated the Aβ accumulation in cerebral vasculature in a dose-dependent manner. Western blot analyses revealed that Pb exposure decreased LRP1 expression in cortical capillaries and hippocampal parenchyma. Immunohistochemistry (IHC) studies further revealed a disrupted distribution of LRP1 alongside hippocampal vasculature accompanied with a decreased expression in hippocampal neurons by Pb exposure. Taken together, the current study demonstrated that the cerebral vasculature naturally possessed a high affinity to Aβ present in circulating blood. Pb exposure significantly increased Aβ accumulation in cerebral vasculature; such an increased Aβ accumulation was due partly to the diminished expression of LRP1 in response to Pb in tested brain regions. Perceivably, Pb-facilitated Ab aggravation in cerebral vasculature may contribute to Pb-associated amyloid alterations.
Collapse
Affiliation(s)
- Luke L. Liu
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907 USA
| | - Xiaoli Shen
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907 USA
- School of Public Health, Qingdao University, Qingdao, China
| | - Huiying Gu
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Gang Zhao
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907 USA
- Department of Medical Biology, School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, China
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, HAMP-1273, West Lafayette, IN 47907 USA
| |
Collapse
|
14
|
Mazura AD, Pietrzik CU. Endocrine Regulation of Microvascular Receptor-Mediated Transcytosis and Its Therapeutic Opportunities: Insights by PCSK9-Mediated Regulation. Pharmaceutics 2023; 15:pharmaceutics15041268. [PMID: 37111752 PMCID: PMC10144601 DOI: 10.3390/pharmaceutics15041268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, many neurological disorders lack effective treatment options due to biological barriers that effectively separate the central nervous system (CNS) from the periphery. CNS homeostasis is maintained by a highly selective exchange of molecules, with tightly controlled ligand-specific transport systems at the blood-brain barrier (BBB) playing a key role. Exploiting or modifying these endogenous transport systems could provide a valuable tool for targeting insufficient drug delivery into the CNS or pathological changes in the microvasculature. However, little is known about how BBB transcytosis is continuously regulated to respond to temporal or chronic changes in the environment. The aim of this mini-review is to draw attention to the sensitivity of the BBB to circulating molecules derived from peripheral tissues, which may indicate a fundamental endocrine-operating regulatory system of receptor-mediated transcytosis at the BBB. We present our thoughts in the context of the recent observation that low-density lipoprotein receptor-related protein 1 (LRP1)-mediated clearance of brain amyloid-β (Aβ) across the BBB is negatively regulated by peripheral proprotein convertase subtilisin/kexin type 9 (PCSK9). We hope that our conclusions will inspire future investigations of the BBB as dynamic communication interface between the CNS and periphery, whose peripheral regulatory mechanisms could be easily exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Alexander D Mazura
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| |
Collapse
|
15
|
Bao H, Shen Y. Unmasking BACE1 in aging and age-related diseases. Trends Mol Med 2023; 29:99-111. [PMID: 36509631 DOI: 10.1016/j.molmed.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022]
Abstract
The beta-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) has long been considered a conventional target for Alzheimer's disease (AD). Unfortunately, AD clinical trials of most BACE1 inhibitors were discontinued due to ineffective cognitive improvement or safety challenges. Recent studies investigating the involvement of BACE1 in metabolic, vascular, and immune functions have indicated a role in aging, diabetes, hypertension, and cancer. These novel BACE1 functions have helped to identify new 'druggable' targets for BACE1 against aging comorbidities. In this review, we discuss BACE1 regulation during aging, and then provide recent insights into its enzymatic and nonenzymatic involvement in aging and age-related diseases. Our study not only proposes the perspective of BACE1's actions in various systems, but also provides new directions for using BACE1 inhibitors and modulators to delay aging and to treat age-related diseases.
Collapse
Affiliation(s)
- Hong Bao
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China; Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Provincial Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 230026, China; CAS Key Laboratory of Brain Function and Disease, Division of Biological and Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
16
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
17
|
Kotlyarov S, Kotlyarova A. Clinical Significance of Lipid Transport Function of ABC Transporters in the Innate Immune System. MEMBRANES 2022; 12:1083. [PMID: 36363640 PMCID: PMC9698216 DOI: 10.3390/membranes12111083] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
ABC transporters are a large family of proteins that transport a variety of substrates across cell plasma membranes. Because of this, they are involved in many physiological processes. It is of interest to note that many ABC transporters are involved in the transport of various lipids. In addition, this function may be related to the innate immune system. The evidence that ABC transporters are involved in the regulation of the innate immune system through the transport of various substances greatly enhances the understanding of their clinical significance. ABC transporters are involved in the cellular homeostasis of cholesterol as well as in the regulation of its content in lipid rafts. Through these mechanisms, they can regulate the function of membrane proteins, including receptors of the innate immune system. By regulating lipid transport, some members of ABC transporters are involved in phagocytosis. In addition, ABC transporters are involved in the transport of lipopolysaccharide, lipid mediators of inflammation, and perform other functions in the innate immune system.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacy Management and Economics, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
18
|
de Mélo Silva Júnior ML, Diniz PRB, de Souza Vilanova MV, Basto GPT, Valença MM. Brain ventricles, CSF and cognition: a narrative review. Psychogeriatrics 2022; 22:544-552. [PMID: 35488797 DOI: 10.1111/psyg.12839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/07/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
The brain ventricles are structures that have been related to cognition since antiquity. They are essential components in the development and maintenance of brain functions. The aging process runs with the enlargement of ventricles and is related to a less selective blood-cerebrospinal fluid barrier and then a more toxic cerebrospinal fluid environment. The study of brain ventricles as a biological marker of aging is promissing because they are structures easily identified in neuroimaging studies, present good inter-rater reliability, and measures of them can identify brain atrophy earlier than cortical structures. The ventricular system also plays roles in the development of dementia, since dysfunction in the clearance of beta-amyloid protein is a key mechanism in sporadic Alzheimer's disease. The morphometric and volumetric studies of the brain ventricles can help to distinguish between healthy elderly and persons with mild cognitive impairment (MCI) and dementia. Brain ventricle data may contribute to the appropriate allocation of individuals in groups at higher risk for MCI-dementia progression in clinical trials and to measuring therapeutic responses in these studies, as well as providing differential diagnosis, such as normal pressure hydrocephalus. Here, we reviewed the pathophysiology of healthy aging and cognitive decline, focusing on the role of the choroid plexus and brain ventricles in this process.
Collapse
Affiliation(s)
- Mário Luciano de Mélo Silva Júnior
- Medical School, Universidade Federal de Pernambuco, Recife, Brazil.,Medical School, Centro Universitário Maurício de Nassau, Recife, Brazil.,Neurology Unit, Hospital da Restauração, Recife, Brazil
| | | | | | | | | |
Collapse
|
19
|
N-terminally truncated Aβ4-x proteoforms and their relevance for Alzheimer's pathophysiology. Transl Neurodegener 2022; 11:30. [PMID: 35641972 PMCID: PMC9158284 DOI: 10.1186/s40035-022-00303-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/21/2022] [Indexed: 01/22/2023] Open
Abstract
Background The molecular heterogeneity of Alzheimer’s amyloid-β (Aβ) deposits extends well beyond the classic Aβ1-40/Aβ1-42 dichotomy, substantially expanded by multiple post-translational modifications that increase the proteome diversity. Numerous truncated fragments consistently populate the brain Aβ peptidome, and their homeostatic regulation and potential contribution to disease pathogenesis are largely unknown. Aβ4-x peptides have been reported as major components of plaque cores and the limited studies available indicate their relative abundance in Alzheimer’s disease (AD). Methods Immunohistochemistry was used to assess the topographic distribution of Aβ4-x species in well-characterized AD cases using custom-generated monoclonal antibody 18H6—specific for Aβ4-x species and blind for full-length Aβ1-40/Aβ1-42—in conjunction with thioflavin-S and antibodies recognizing Aβx-40 and Aβx-42 proteoforms. Circular dichroism, thioflavin-T binding, and electron microscopy evaluated the biophysical and aggregation/oligomerization properties of full-length and truncated synthetic homologues, whereas stereotaxic intracerebral injections of monomeric and oligomeric radiolabeled homologues in wild-type mice were used to evaluate their brain clearance characteristics. Results All types of amyloid deposits contained the probed Aβ epitopes, albeit expressed in different proportions. Aβ4-x species showed preferential localization within thioflavin-S-positive cerebral amyloid angiopathy and cored plaques, strongly suggesting poor clearance characteristics and consistent with the reduced solubility and enhanced oligomerization of their synthetic homologues. In vivo clearance studies demonstrated a fast brain efflux of N-terminally truncated and full-length monomeric forms whereas their oligomeric counterparts—particularly of Aβ4-40 and Aβ4-42—consistently exhibited enhanced brain retention. Conclusions The persistence of aggregation-prone Aβ4-x proteoforms likely contributes to the process of amyloid formation, self-perpetuating the amyloidogenic loop and exacerbating amyloid-mediated pathogenic pathways.
Collapse
|
20
|
Leite DM, Seifi M, Ruiz-Perez L, Nguemo F, Plomann M, Swinny JD, Battaglia G. Syndapin-2 mediated transcytosis of amyloid-ß across the blood-brain barrier. Brain Commun 2022; 4:fcac039. [PMID: 35233527 PMCID: PMC8882007 DOI: 10.1093/braincomms/fcac039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/31/2021] [Accepted: 02/15/2022] [Indexed: 11/14/2022] Open
Abstract
A deficient transport of amyloid-β across the blood–brain barrier, and its diminished clearance from the brain, contribute to neurodegenerative and vascular pathologies, such as Alzheimer’s disease and cerebral amyloid angiopathy, respectively. At the blood–brain barrier, amyloid-β efflux transport is associated with the low-density lipoprotein receptor-related protein 1. However, the precise mechanisms governing amyloid-β transport across the blood–brain barrier, in health and disease, remain to be fully understood. Recent evidence indicates that the low-density lipoprotein receptor-related protein 1 transcytosis occurs through a tubulation-mediated mechanism stabilized by syndapin-2. Here, we show that syndapin-2 is associated with amyloid-β clearance via low-density lipoprotein receptor-related protein 1 across the blood–brain barrier. We further demonstrate that risk factors for Alzheimer’s disease, amyloid-β expression and ageing, are associated with a decline in the native expression of syndapin-2 within the brain endothelium. Our data reveals that syndapin-2-mediated pathway, and its balance with the endosomal sorting, are important for amyloid-β clearance proposing a measure to evaluate Alzheimer’s disease and ageing, as well as a target for counteracting amyloid-β build-up. Moreover, we provide evidence for the impact of the avidity of amyloid-β assemblies in their trafficking across the brain endothelium and in low-density lipoprotein receptor-related protein 1 expression levels, which may affect the overall clearance of amyloid-β across the blood–brain barrier.
Collapse
Affiliation(s)
- Diana M. Leite
- Department of Chemistry, University College London, London, United Kingdom
- Institute for the Physics of Living Systems, University College London, London, United Kingdom
| | - Mohsen Seifi
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Lorena Ruiz-Perez
- Department of Chemistry, University College London, London, United Kingdom
- Institute for the Physics of Living Systems, University College London, London, United Kingdom
| | - Filomain Nguemo
- Institute for Neurophysiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Markus Plomann
- Institute of Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jerome D. Swinny
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, United Kingdom
| | - Giuseppe Battaglia
- Department of Chemistry, University College London, London, United Kingdom
- Institute for the Physics of Living Systems, University College London, London, United Kingdom
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
21
|
Elbert DL, Patterson BW, Lucey BP, Benzinger TLS, Bateman RJ. Importance of CSF-based Aβ clearance with age in humans increases with declining efficacy of blood-brain barrier/proteolytic pathways. Commun Biol 2022; 5:98. [PMID: 35087179 PMCID: PMC8795390 DOI: 10.1038/s42003-022-03037-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022] Open
Abstract
The kinetics of amyloid beta turnover within human brain is still poorly understood. We previously found a dramatic decline in the turnover of Aβ peptides in normal aging. It was not known if brain interstitial fluid/cerebrospinal fluid (ISF/CSF) fluid exchange, CSF turnover, blood-brain barrier function or proteolysis were affected by aging or the presence of β amyloid plaques. Here, we describe a non-steady state physiological model developed to decouple CSF fluid transport from other processes. Kinetic parameters were estimated using: (1) MRI-derived brain volumes, (2) stable isotope labeling kinetics (SILK) of amyloid-β peptide (Aβ), and (3) lumbar CSF Aβ concentration during SILK. Here we show that changes in blood-brain barrier transport and/or proteolysis were largely responsible for the age-related decline in Aβ turnover rates. CSF-based clearance declined modestly in normal aging but became increasingly important due to the slowing of other processes. The magnitude of CSF-based clearance was also lower than that due to blood-brain barrier function plus proteolysis. These results suggest important roles for blood-brain barrier transport and proteolytic degradation of Aβ in the development Alzheimer’s Disease in humans. To understand if brain interstitial fluid/cerebrospinal fluid (ISF/CSF) exchange, CSF turnover, blood-brain barrier function or proteolysis were affected by aging or the presence of β amyloid plaques, Elbert et al. develop a non-steady state physiological model using MRI-derived brain volumes, stable isotope labeling kinetics of Aβ, and lumbar CSF Aβ concentration. Their model suggests an important role for blood-brain barrier transport and proteolytic degradation of Aβ in the development Alzheimer’s Disease in humans.
Collapse
Affiliation(s)
- Donald L Elbert
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
| | - Bruce W Patterson
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Brendan P Lucey
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie L S Benzinger
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA.,Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
22
|
Gião T, Teixeira T, Almeida MR, Cardoso I. Choroid Plexus in Alzheimer's Disease-The Current State of Knowledge. Biomedicines 2022; 10:224. [PMID: 35203434 PMCID: PMC8869376 DOI: 10.3390/biomedicines10020224] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 01/31/2023] Open
Abstract
The choroid plexus (CP), located in each of the four ventricles of the brain, is formed by a monolayer of epithelial cells that surrounds a highly vascularized connective tissue with permeable capillaries. These cells are joined by tight junctions forming the blood-cerebrospinal fluid barrier (BCSFB), which strictly regulates the exchange of substances between the blood and cerebrospinal fluid (CSF). The primary purpose of the CP is to secrete CSF, but it also plays a role in the immune surveillance of the central nervous system (CNS) and in the removal of neurotoxic compounds from the CSF. According to recent findings, the CP is also involved in the modulation of the circadian cycle and neurogenesis. In diseases such as Alzheimer's disease (AD), the function of the CP is impaired, resulting in an altered secretory, barrier, transport, and immune function. This review describes the current state of knowledge concerning the roles of the CP and BCSFB in the pathophysiology of AD and summarizes recently proposed therapies that aim to restore CP and BCSFB functions.
Collapse
Affiliation(s)
- Tiago Gião
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| | - Tiago Teixeira
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Maria Rosário Almeida
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| | - Isabel Cardoso
- Molecular Neurobiology Group, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (T.T.); (M.R.A.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS—Instituto de Ciências Biomédicas Abel Salazar, 4050-013 Porto, Portugal
| |
Collapse
|
23
|
Zhou AL, Sharda N, Sarma VV, Ahlschwede KM, Curran GL, Tang X, Poduslo JF, Kalari KR, Lowe VJ, Kandimalla KK. Age-Dependent Changes in the Plasma and Brain Pharmacokinetics of Amyloid-β Peptides and Insulin. J Alzheimers Dis 2022; 85:1031-1044. [PMID: 34924382 PMCID: PMC10846947 DOI: 10.3233/jad-215128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Age is the most common risk factor for Alzheimer's disease (AD), a neurodegenerative disorder characterized by the hallmarks of toxic amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. Moreover, sub-physiological brain insulin levels have emerged as a pathological manifestation of AD. OBJECTIVE Identify age-related changes in the plasma disposition and blood-brain barrier (BBB) trafficking of Aβ peptides and insulin in mice. METHODS Upon systemic injection of 125I-Aβ40, 125I-Aβ42, or 125I-insulin, the plasma pharmacokinetics and brain influx were assessed in wild-type (WT) or AD transgenic (APP/PS1) mice at various ages. Additionally, publicly available single-cell RNA-Seq data [GSE129788] was employed to investigate pathways regulating BBB transport in WT mice at different ages. RESULTS The brain influx of 125I-Aβ40, estimated as the permeability-surface area product, decreased with age, accompanied by an increase in plasma AUC. In contrast, the brain influx of 125I-Aβ42 increased with age, accompanied by a decrease in plasma AUC. The age-dependent changes observed in WT mice were accelerated in APP/PS1 mice. As seen with 125I-Aβ40, the brain influx of 125I-insulin decreased with age in WT mice, accompanied by an increase in plasma AUC. This finding was further supported by dynamic single-photon emission computed tomography (SPECT/CT) imaging studies. RAGE and PI3K/AKT signaling pathways at the BBB, which are implicated in Aβ and insulin transcytosis, respectively, were upregulated with age in WT mice, indicating BBB insulin resistance. CONCLUSION Aging differentially affects the plasma pharmacokinetics and brain influx of Aβ isoforms and insulin in a manner that could potentially augment AD risk.
Collapse
Affiliation(s)
- Andrew L. Zhou
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Nidhi Sharda
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Vidur V. Sarma
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Kristen M. Ahlschwede
- Department of Pharmaceutical Sciences, Rosalind Franklin University of Medicine and Science, College of Pharmacy, North Chicago, IL, USA
| | - Geoffry L. Curran
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Xiaojia Tang
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Joseph F. Poduslo
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Krishna R. Kalari
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Karunya K. Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| |
Collapse
|
24
|
Abstract
The accumulation of neurotoxic amyloid-beta (Aβ) in the brain is one of the characteristic hallmarks of Alzheimer's disease (AD). Aβ-peptide brain homeostasis is governed by its production and various clearance mechanisms. The blood-brain barrier provides a large surface area for influx and efflux mechanisms into and out of the brain. Different transporters and receptors have been implicated to play crucial roles in Aβ clearance from brain. Besides Aβ transport, the blood-brain barrier tightly regulates the brain's microenvironment; however, vascular alterations have been shown in patients with AD. Here, we summarize how the blood-brain barrier changes during aging and in disease and focus on recent findings of how the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) play a role in Aβ clearance from brain.
Collapse
|
25
|
Yuan M, Wang Y, Wang S, Huang Z, Jin F, Zou Q, Li J, Pu Y, Cai Z. Bioenergetic Impairment in the Neuro-Glia-Vascular Unit: An Emerging Physiopathology during Aging. Aging Dis 2021; 12:2080-2095. [PMID: 34881087 PMCID: PMC8612602 DOI: 10.14336/ad.2021.04017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
An emerging concept termed the "neuro-glia-vascular unit" (NGVU) has been established in recent years to understand the complicated mechanism of multicellular interactions among vascular cells, glial cells, and neurons. It has been proverbially reported that the NGVU is significantly associated with neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Physiological aging is an inevitable progression associated with oxidative damage, bioenergetic alterations, mitochondrial dysfunction, and neuroinflammation, which is partially similar to the pathology of AD. Thus, senescence is regarded as the background for the development of neurodegenerative diseases. With the exacerbation of global aging, senescence is an increasingly serious problem in the medical field. In this review, the coupling of each component, including neurons, glial cells, and vascular cells, in the NGVU is described in detail. Then, various mechanisms of age-dependent impairment in each part of the NGVU are discussed. Moreover, the potential bioenergetic alterations between different cell types in the NGVU are highlighted, which seems to be an emerging physiopathology associated with the aged brain. Bioenergetic intervention in the NGVU may be a new direction for studies on delaying or diminishing aging in the future.
Collapse
Affiliation(s)
- Minghao Yuan
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Yangyang Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Shengyuan Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Zhenting Huang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Feng Jin
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Qian Zou
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Jing Li
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Yinshuang Pu
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Zhiyou Cai
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Nguyen B, Bix G, Yao Y. Basal lamina changes in neurodegenerative disorders. Mol Neurodegener 2021; 16:81. [PMID: 34876200 PMCID: PMC8650282 DOI: 10.1186/s13024-021-00502-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neurodegenerative disorders are a group of age-associated diseases characterized by progressive degeneration of the structure and function of the CNS. Two key pathological features of these disorders are blood-brain barrier (BBB) breakdown and protein aggregation. MAIN BODY The BBB is composed of various cell types and a non-cellular component---the basal lamina (BL). Although how different cells affect the BBB is well studied, the roles of the BL in BBB maintenance and function remain largely unknown. In addition, located in the perivascular space, the BL is also speculated to regulate protein clearance via the meningeal lymphatic/glymphatic system. Recent studies from our laboratory and others have shown that the BL actively regulates BBB integrity and meningeal lymphatic/glymphatic function in both physiological and pathological conditions, suggesting that it may play an important role in the pathogenesis and/or progression of neurodegenerative disorders. In this review, we focus on changes of the BL and its major components during aging and in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). First, we introduce the vascular and lymphatic systems in the CNS. Next, we discuss the BL and its major components under homeostatic conditions, and summarize their changes during aging and in AD, PD, and ALS in both rodents and humans. The functional significance of these alterations and potential therapeutic targets are also reviewed. Finally, key challenges in the field and future directions are discussed. CONCLUSIONS Understanding BL changes and the functional significance of these changes in neurodegenerative disorders will fill the gap of knowledge in the field. Our goal is to provide a clear and concise review of the complex relationship between the BL and neurodegenerative disorders to stimulate new hypotheses and further research in this field.
Collapse
Affiliation(s)
- Benjamin Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Gregory Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA.
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, Tampa, Florida, 33612, USA.
| |
Collapse
|
27
|
Gireud-Goss M, Mack AF, McCullough LD, Urayama A. Cerebral Amyloid Angiopathy and Blood-Brain Barrier Dysfunction. Neuroscientist 2021; 27:668-684. [PMID: 33238806 PMCID: PMC9853919 DOI: 10.1177/1073858420954811] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cerebral hemorrhage, a devastating subtype of stroke, is often caused by hypertension and cerebral amyloid angiopathy (CAA). Pathological evidence of CAA is detected in approximately half of all individuals over the age of 70 and is associated with cortical microinfarcts and cognitive impairment. The underlying pathophysiology of CAA is characterized by accumulation of pathogenic amyloid β (Aβ) fragments of amyloid precursor protein in the cerebral vasculature. Vascular deposition of Aβ damages the vessel wall, results in blood-brain barrier (BBB) leakiness, vessel occlusion or rupture, and leads to hemorrhages and decreased cerebral blood flow that negatively affects vessel integrity and cognitive function. Currently, the main hypothesis surrounding the mechanism of CAA pathogenesis is that there is an impaired clearance of Aβ peptides, which includes compromised perivascular drainage as well as dysfunction of BBB transport. Also, the immune response in CAA pathogenesis plays an important role. Therefore, the mechanism by which Aβ vascular deposition occurs is crucial for our understanding of CAA pathogenesis and for the development of potential therapeutic options.
Collapse
Affiliation(s)
- Monica Gireud-Goss
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexis F. Mack
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
28
|
Bubak AN, Como CN, Hassell JE, Mescher T, Frietze SE, Niemeyer CS, Cohrs RJ, Nagel MA. Targeted RNA Sequencing of VZV-Infected Brain Vascular Adventitial Fibroblasts Indicates That Amyloid May Be Involved in VZV Vasculopathy. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 9:9/1/e1103. [PMID: 34759019 PMCID: PMC8587729 DOI: 10.1212/nxi.0000000000001103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/09/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVES Compared with stroke controls, patients with varicella zoster virus (VZV) vasculopathy have increased amyloid in CSF, along with increased amylin (islet amyloid polypeptide [IAPP]) and anti-VZV antibodies. Thus, we examined the gene expression profiles of VZV-infected primary human brain vascular adventitial fibroblasts (HBVAFs), one of the initial arterial cells infected in VZV vasculopathy, to determine whether they are a potential source of amyloid that can disrupt vasculature and potentiate inflammation. METHODS Mock- and VZV-infected quiescent HBVAFs were harvested at 3 days postinfection. Targeted RNA sequencing of the whole-human transcriptome (BioSpyder Technologies, TempO-Seq) was conducted followed by gene set enrichment and pathway analysis. Selected pathways unique to VZV-infected cells were confirmed by enzyme-linked immunoassays, migration assays, and immunofluorescence analysis (IFA) that included antibodies against amylin and amyloid-beta, as well as amyloid staining by Thioflavin-T. RESULTS Compared with mock, VZV-infected HBVAFs had significantly enriched gene expression pathways involved in vascular remodeling and vascular diseases; confirmatory studies showed secretion of matrix metalloproteinase-3 and -10, as well increased migration of infected cells and uninfected cells when exposed to conditioned media from VZV-infected cells. In addition, significantly enriched pathways involved in amyloid-associated diseases (diabetes mellitus, amyloidosis, and Alzheimer disease), tauopathy, and progressive neurologic disorder were identified; predicted upstream regulators included amyloid precursor protein, apolipoprotein E, microtubule-associated protein tau, presenilin 1, and IAPP. Confirmatory IFA showed that VZV-infected HBVAFs contained amyloidogenic peptides (amyloid-beta and amylin) and intracellular amyloid. DISCUSSION Gene expression profiles and pathway enrichment analysis of VZV-infected HBVAFs, as well as phenotypic studies, reveal features of pathologic vascular remodeling (e.g., increased cell migration and changes in the extracellular matrix) that can contribute to cerebrovascular disease. Furthermore, the discovery of amyloid-associated transcriptional pathways and intracellular amyloid deposition in HBVAFs raise the possibility that VZV vasculopathy is an amyloid disease. Amyloid deposition may contribute to cell death and loss of vascular wall integrity, as well as potentiate chronic inflammation in VZV vasculopathy, with disease severity and recurrence determined by the host's ability to clear virus infection and amyloid deposition and by the coexistence of other amyloid-associated diseases (i.e., Alzheimer disease and diabetes mellitus).
Collapse
Affiliation(s)
- Andrew N Bubak
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Christina N Como
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - James E Hassell
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Teresa Mescher
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Seth E Frietze
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Christy S Niemeyer
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Randall J Cohrs
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO
| | - Maria A Nagel
- From the Department of Neurology (A.N.B., C.N.C., J.E.H., T.M., C.S.N., R.J.C., M.A.N.), University of Colorado; Department of Medical Laboratory Sciences (S.E.F.), University of Vermont, Burlington, VT; Department of Immununology & Microbiology (R.J.C.), University of Colorado; and Department of Ophthalmology (M.A.N.), University of Colorado, Aurora, CO.
| |
Collapse
|
29
|
Vandendriessche C, Balusu S, Van Cauwenberghe C, Brkic M, Pauwels M, Plehiers N, Bruggeman A, Dujardin P, Van Imschoot G, Van Wonterghem E, Hendrix A, Baeke F, De Rycke R, Gevaert K, Vandenbroucke RE. Importance of extracellular vesicle secretion at the blood-cerebrospinal fluid interface in the pathogenesis of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:143. [PMID: 34425919 PMCID: PMC8381545 DOI: 10.1186/s40478-021-01245-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence indicates that extracellular vesicles (EVs) play an important role in the pathogenesis of Alzheimer's disease (AD). We previously reported that the blood-cerebrospinal fluid (CSF) interface, formed by the choroid plexus epithelial (CPE) cells, releases an increased amount of EVs into the CSF in response to peripheral inflammation. Here, we studied the importance of CP-mediated EV release in AD pathogenesis. We observed increased EV levels in the CSF of young transgenic APP/PS1 mice which correlated with high amyloid beta (Aβ) CSF levels at this age. The intracerebroventricular (icv) injection of Aβ oligomers (AβO) in wild-type mice revealed a significant increase of EVs in the CSF, signifying that the presence of CSF-AβO is sufficient to induce increased EV secretion. Using in vivo, in vitro and ex vivo approaches, we identified the CP as a major source of the CSF-EVs. Interestingly, AβO-induced, CP-derived EVs induced pro-inflammatory effects in mixed cortical cultures. Proteome analysis of these EVs revealed the presence of several pro-inflammatory proteins, including the complement protein C3. Strikingly, inhibition of EV production using GW4869 resulted in protection against acute AβO-induced cognitive decline. Further research into the underlying mechanisms of this EV secretion might open up novel therapeutic strategies to impact the pathogenesis and progression of AD.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sriram Balusu
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
| | - Caroline Van Cauwenberghe
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marjana Brkic
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Marie Pauwels
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nele Plehiers
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Femke Baeke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB BioImaging Core, Ghent, Belgium
| | - Riet De Rycke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB BioImaging Core, Ghent, Belgium
| | - Kris Gevaert
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
30
|
Peng HB, Bukuroshi P, Durk MR, Grootendorst P, Yan X, Pan SR, de Lannoy IAM, Pang KS. Impact of age, hypercholesterolemia, and the vitamin D receptor on brain endogenous β-amyloid peptide accumulation in mice. Biopharm Drug Dispos 2021; 42:372-388. [PMID: 34219248 DOI: 10.1002/bdd.2297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
Age, hypercholesterolemia, and vitamin D deficiency are risk factors that increase the brain accumulation of pathogenic β-amyloid peptides (40 and 42), precursors leading to Alzheimer's disease (AD) in humans. The relative changes accompanying aging, high cholesterol, and/or treatment of calcitriol, active vitamin D receptor (VDR) ligand, under normal physiology are unknown. We examined these relative changes in C57BL/6 mice of ages 2, 4-8, and more than 10 months old, which were fed a normal or high fat / high cholesterol diet and treated with calcitriol, active ligand of the vitamin D receptor (0 or 2.5 μg/kg ×4, intraperitoneally, every other day to elicit cholesterol lowering in liver). Aβ40 but not Aβ42 accumulation in brain and lower P-glycoprotein (P-gp) and neprilysin protein expressions for Aβ efflux and degradation, respectively, were found to be associated with aging. But there was no trend for BACE1 (β-secretase 1, a cholesterol-sensitive enzyme) toward Aβ synthesis with age. In response to calcitriol treatment, P-gp was elevated, mitigating partially the age-related changes. Although age-dependent decreasing trends in mRNA expression levels existed for Cyp46a1, the brain cholesterol processing enzyme, whose inhibition increases BACE1 and ApoE to facilitate microglia Aβ degradation, mRNA changes for other cholesterol transporters: Acat1 and Abca1, and brain cholesterol levels remained unchanged. There was no observable change in the mRNA expression of amyloid precursor protein (APP) and the influx (RAGE) and efflux (LRP1) transporters with respect to age, diet, or calcitriol treatment. Overall, aging poses as a risk factor contributing to Aβ accumulation in brain, and VDR-mediated P-gp activation partially alleviates the outcome.
Collapse
Affiliation(s)
- H Benson Peng
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paola Bukuroshi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Matthew R Durk
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paul Grootendorst
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoyu Yan
- Department of Pharmacy, Chinese University of Hong Kong, Hong Kong, China
| | - Sophie R Pan
- InterVivo Solutions Inc., Mississauga, Ontario, Canada
| | | | - K Sandy Pang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Mather M. Noradrenaline in the aging brain: Promoting cognitive reserve or accelerating Alzheimer's disease? Semin Cell Dev Biol 2021; 116:108-124. [PMID: 34099360 PMCID: PMC8292227 DOI: 10.1016/j.semcdb.2021.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
Many believe that engaging in novel and mentally challenging activities promotes brain health and prevents Alzheimer's disease in later life. However, mental stimulation may also have risks as well as benefits. As neurons release neurotransmitters, they often also release amyloid peptides and tau proteins into the extracellular space. These by-products of neural activity can aggregate into the tau tangle and amyloid plaque signatures of Alzheimer's disease. Over time, more active brain regions accumulate more pathology. Thus, increasing brain activity can have a cost. But the neuromodulator noradrenaline, released during novel and mentally stimulating events, may have some protective effects-as well as some negative effects. Via its inhibitory and excitatory effects on neurons and microglia, noradrenaline sometimes prevents and sometimes accelerates the production and accumulation of amyloid-β and tau in various brain regions. Both α2A- and β-adrenergic receptors influence amyloid-β production and tau hyperphosphorylation. Adrenergic activity also influences clearance of amyloid-β and tau. Furthermore, some findings suggest that Alzheimer's disease increases noradrenergic activity, at least in its early phases. Because older brains clear the by-products of synaptic activity less effectively, increased synaptic activity in the older brain risks accelerating the accumulation of Alzheimer's pathology more than it does in the younger brain.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, & Department of Biomedical Engineering, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089, United States.
| |
Collapse
|
32
|
Liu L, Ni YQ, Zhan JK, Liu YS. The Role of SGLT2 Inhibitors in Vascular Aging. Aging Dis 2021; 12:1323-1336. [PMID: 34341711 PMCID: PMC8279525 DOI: 10.14336/ad.2020.1229] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular aging is defined as organic and functional changes in blood vessels, in which decline in autophagy levels, DNA damage, MicroRNA (miRNA), oxidative stress, sirtuin, and apoptosis signal-regulated kinase 1 (ASK1) are integral thereto. With regard to vascular morphology, the increase in arterial stiffness, atherosclerosis, vascular calcification and high amyloid beta levels are closely related to vascular aging. Further closely related thereto, at the cellular level, is the aging of vascular endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). Vascular aging seriously affects the health, economy and life of patients, but can be delayed by SGLT2 inhibitors through the improvement of vascular function. In the present article, a review is conducted of recent domestic and international progress in research on SGLT2 inhibitors,vascular aging and diseases related thereto, thereby providing theoretical support and guidance for further revealing the relationship between SGLT2 inhibitors and diseases related to vascular aging.
Collapse
Affiliation(s)
- Le Liu
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yu-Qing Ni
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Jun-Kun Zhan
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - You-Shuo Liu
- 1Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,2Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
33
|
das Neves SP, Delivanoglou N, Da Mesquita S. CNS-Draining Meningeal Lymphatic Vasculature: Roles, Conundrums and Future Challenges. Front Pharmacol 2021; 12:655052. [PMID: 33995074 PMCID: PMC8113819 DOI: 10.3389/fphar.2021.655052] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
A genuine and functional lymphatic vascular system is found in the meninges that sheath the central nervous system (CNS). This unexpected (re)discovery led to a reevaluation of CNS fluid and solute drainage mechanisms, neuroimmune interactions and the involvement of meningeal lymphatics in the initiation and progression of neurological disorders. In this manuscript, we provide an overview of the development, morphology and unique functional features of meningeal lymphatics. An outline of the different factors that affect meningeal lymphatic function, such as growth factor signaling and aging, and their impact on the continuous drainage of brain-derived molecules and meningeal immune cells into the cervical lymph nodes is also provided. We also highlight the most recent discoveries about the roles of the CNS-draining lymphatic vasculature in different pathologies that have a strong neuroinflammatory component, including brain trauma, tumors, and aging-associated neurodegenerative diseases like Alzheimer's and Parkinson's. Lastly, we provide a critical appraisal of the conundrums, challenges and exciting questions involving the meningeal lymphatic system that ought to be investigated in years to come.
Collapse
Affiliation(s)
| | | | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
34
|
Abuelezz SA, Hendawy N. HMGB1/RAGE/TLR4 axis and glutamate as novel targets for PCSK9 inhibitor in high fat cholesterol diet induced cognitive impairment and amyloidosis. Life Sci 2021; 273:119310. [PMID: 33667517 DOI: 10.1016/j.lfs.2021.119310] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 11/15/2022]
Abstract
AIMS Alzheimer's disease (AD) is a leading health problem in which increased amyloid β (Aβ) accumulation may occur due to abnormal Aβ precursor protein processing by β-secretase 1 (BACE1) enzyme. Lately, neuro-inflammation was recognized as a significant contributor to its pathogenesis. Although the causes of AD are not yet well understood, much evidence has suggested that dyslipidemia has harmful effects on cognitive function and is inextricably involved in AD pathogenesis. Cholesterol is a vital molecule involved in neuronal development. Alteration in neuronal cholesterol levels affects Aβ metabolism and results in neurodegeneration. Proprotein-convertase-subtilisin/kexin type-9 (PCSK9) was found to decrease neuronal cholesterol uptake by degradation of LDL-receptor related protein 1 (LRP-1) responsible for neuronal cholesterol uptake. Accordingly, this study was designed to evaluate the effect of PCSK9-inhibition by alirocumab (Aliro) in high-fat-cholesterol-diet (HFCD)-induced-AD-like condition. MAIN METHODS Wistar Rats were divided into six groups; control; HFCD; HFCD and Memantine; HFCD and Aliro (4, 8 and 16 mg/kg/week) to test for ability of Aliro to modulate cognitive impairment, amyloidosis, brain cholesterol homeostasis and neuro-inflammation in HFCD-induced-AD-like condition. KEY FINDINGS Our results demonstrated an association between PCSK9 inhibition by Aliro and amelioration of cognitive deficit, cholesterol hemostasis and reduction of neuro-inflammation. Aliro was able to alleviate hippocampal LRP-1expression levels and reduce brain cholesterol, hippocampal BACE1, Aβ42, high-mobility-group-box-1 protein, receptor for advanced-glycation-end-products and toll like receptor-4 with subsequent decrease of different inflammatory mediators as nuclear-factor-kappa-B (NF-κB), tumor-necrosis-factor-alpha (TNF-α), interleukin-1beta (IL-1β) and IL-6. SIGNIFICANCE PCSK9-inhibition may represent a new therapeutic target in AD especially for HFCD-induced-AD-like condition.
Collapse
Affiliation(s)
- Sally A Abuelezz
- Clinical Pharmacology Department, Faculty of Medicine Ain-Shams University, Cairo, Egypt.
| | - Nevien Hendawy
- Clinical Pharmacology Department, Faculty of Medicine Ain-Shams University, Cairo, Egypt
| |
Collapse
|
35
|
Steinman J, Sun HS, Feng ZP. Microvascular Alterations in Alzheimer's Disease. Front Cell Neurosci 2021; 14:618986. [PMID: 33536876 PMCID: PMC7849053 DOI: 10.3389/fncel.2020.618986] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with continual decline in cognition and ability to perform routine functions such as remembering familiar places or understanding speech. For decades, amyloid beta (Aβ) was viewed as the driver of AD, triggering neurodegenerative processes such as inflammation and formation of neurofibrillary tangles (NFTs). This approach has not yielded therapeutics that cure the disease or significant improvements in long-term cognition through removal of plaques and Aβ oligomers. Some researchers propose alternate mechanisms that drive AD or act in conjunction with amyloid to promote neurodegeneration. This review summarizes the status of AD research and examines research directions including and beyond Aβ, such as tau, inflammation, and protein clearance mechanisms. The effect of aging on microvasculature is highlighted, including its contribution to reduced blood flow that impairs cognition. Microvascular alterations observed in AD are outlined, emphasizing imaging studies of capillary malfunction. The review concludes with a discussion of two therapies to protect tissue without directly targeting Aβ for removal: (1) administration of growth factors to promote vascular recovery in AD; (2) inhibiting activity of a calcium-permeable ion channels to reduce microglial activation and restore cerebral vascular function.
Collapse
Affiliation(s)
- Joe Steinman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Hsu HW, Rodriguez-Ortiz CJ, Zumkehr J, Kitazawa M. Inflammatory Cytokine IL-1β Downregulates Endothelial LRP1 via MicroRNA-mediated Gene Silencing. Neuroscience 2021; 453:69-80. [PMID: 33246059 PMCID: PMC7796931 DOI: 10.1016/j.neuroscience.2020.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/26/2022]
Abstract
Effective clearance of neurotoxic amyloid-beta (Aβ) from the brain is a critical process to prevent Alzheimer's disease (AD). One major clearance mechanism is Aβ transcytosis mediated by low-density lipoprotein receptor-related protein 1 (LRP1) in capillary endothelial cells. A marked loss of endothelial LRP1 is found in AD brains and is believed to significantly impair Aβ clearance. Recently, we demonstrated that pro-inflammatory cytokines IL-1β, IL-6 and TNF-α, significantly down-regulated LRP1 in human primary microvascular endothelial cells (MVECs). In this study, we sought to determine the underlying molecular mechanism by which IL-1β led to LRP1 loss in MVECs. Reduced LRP1 protein and transcript were detected up to 24 h post-exposure and returned to the baseline levels after 48 h post-exposure with 1 ng/ml IL-1β. This reduction was in part mediated by microRNA-205-5p, -200b-3p, and -200c-3p, as these microRNAs were concomitantly upregulated in MVECs exposed to IL-1β. Synthetic microRNA-205-5p, -200b-3p, and -200c-3p mimics recapitulated LRP1 loss in MVECs without IL-1β, and their synthetic antagomirs effectively reversed IL-1β-mediated LRP1 loss. Importantly, we found that the expression of these three microRNAs was controlled by NF-κB as pharmacological NF-κB inhibitor, BMS-345541, inhibited the IL-1β-mediated upregulation of these microRNAs and rescued LRP1 expression. siRNA-mediated silencing of IκB in MVECs elevated microRNA-200b-3p and decreased LRP1 transcript, partially confirming our overall findings. In conclusion, our study provides a mechanism by which pro-inflammatory IL-1β instigates the suppression of LRP1 expression in MVECs. Our findings could implicate spatiotemporal loss of LRP1 and impairment of the LRP1-mediated clearance mechanism by endothelial cells.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Joannee Zumkehr
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, Department of Environmental and Occupational Health and Department of Medicine, University of California, Irvine, CA, United States.
| |
Collapse
|
37
|
Bouji M, Lecomte A, Gamez C, Blazy K, Villégier AS. Impact of Cerebral Radiofrequency Exposures on Oxidative Stress and Corticosterone in a Rat Model of Alzheimer's Disease. J Alzheimers Dis 2020; 73:467-476. [PMID: 31796670 DOI: 10.3233/jad-190593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common type of neurodegenerative disease leading to dementia. Several studies suggested that mobile phone radiofrequency electromagnetic field (RF-EMF) exposures modified AD memory deficits in rodent models. OBJECTIVE Here we aimed to test the hypothesis that RF-EMF exposure may modify memory through corticosterone and oxidative stress in the Samaritan rat model of AD. METHODS Long-Evans male rats received intracerebroventricular infusion with ferrous sulphate, amyloid-beta 1-42 peptide, and buthionine-sufloximine (AD rats) or with vehicle (control rats). To mimic cell phone use, RF-EMF were exposed to the head for 1 month (5 days/week, in restraint). To look for hazard thresholds, high brain averaged specific absorption rates (BASAR) were tested: 1.5 W/Kg (15 min), 6 W/Kg (15 min), and 6 W/Kg (45 min). The sham group was in restraint for 45 min. Endpoints were spatial memory in the radial maze, plasmatic corticosterone, heme oxygenase-1 (HO1), and amyloid plaques. RESULTS Results indicated similar corticosterone levels but impaired memory performances and increased cerebral staining of thioflavine and of HO1 in the sham AD rats compared to the controls. A correlative increase of cortical HO1 staining was the only effect of RF-EMF in control rats. In AD rats, RF-EMF exposures induced a correlative increase of hippocampal HO1 staining and reduced corticosterone. DISCUSSION According to our data, neither AD nor control rats showed modified memory after RF-EMF exposures. Unlike control rats, AD rats showed higher hippocampal oxidative stress and reduced corticosterone with the higher BASAR. This data suggests more fragility related to neurodegenerative disease toward RF-EMF exposures.
Collapse
Affiliation(s)
- Marc Bouji
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France
| | - Anthony Lecomte
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Christelle Gamez
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Kelly Blazy
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| | - Anne-Sophie Villégier
- Unité de Toxicologie Expérimentale, Institut National de l'Environnement Industriel et des Risques (INERIS), Parc Technologique ALATA, Verneuil-en-Halatte, France.,PERITOX UMR I-01 INERIS 01 UFR de médecine, Amiens, France
| |
Collapse
|
38
|
Watanabe C, Imaizumi T, Kawai H, Suda K, Honma Y, Ichihashi M, Ema M, Mizutani KI. Aging of the Vascular System and Neural Diseases. Front Aging Neurosci 2020; 12:557384. [PMID: 33132896 PMCID: PMC7550630 DOI: 10.3389/fnagi.2020.557384] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022] Open
Abstract
Vertebrates have acquired complex high-order functions facilitated by the dispersion of vascular and neural networks to every corner of the body. Blood vessels deliver oxygen and nutrients to all cells and provide essential transport systems for removing waste products. For these functions, tissue vascularization must be spatiotemporally appropriate. Recent studies revealed that blood vessels create a tissue-specific niche, thus attracting attention as biologically active sites for tissue development. Each capillary network is critical for maintaining proper brain function because age-related and disease-related impairment of cognitive function is associated with the loss or diminishment of brain capillaries. This review article highlights how structural and functional alterations in the brain vessels may change with age and neurogenerative diseases. Capillaries are also responsible for filtering toxic byproducts, providing an appropriate vascular environment for neuronal function. Accumulation of amyloid β is a key event in Alzheimer’s disease pathogenesis. Recent studies have focused on associations reported between Alzheimer’s disease and vascular aging. Furthermore, the glymphatic system and meningeal lymphatic systems contribute to a functional unit for clearance of amyloid β from the brain from the central nervous system into the cervical lymph nodes. This review article will also focus on recent advances in stem cell therapies that aim at repopulation or regeneration of a degenerating vascular system for neural diseases.
Collapse
Affiliation(s)
- Chisato Watanabe
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan.,Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan
| | - Tsutomu Imaizumi
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Hiromi Kawai
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Kazuma Suda
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Yoichi Honma
- Basic Research Development Division, Rohto Pharmaceutical Co., Ltd., Osaka, Japan
| | - Masamitsu Ichihashi
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University Institute for Advanced Study, Kyoto, Japan
| | - Ken-Ichi Mizutani
- Laboratory of Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
39
|
Liu XY, Yang LP, Zhao L. Stem cell therapy for Alzheimer's disease. World J Stem Cells 2020; 12:787-802. [PMID: 32952859 PMCID: PMC7477654 DOI: 10.4252/wjsc.v12.i8.787] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive impairment. It is caused by synaptic failure and excessive accumulation of misfolded proteins. To date, almost all advanced clinical trials on specific AD-related pathways have failed mostly due to a large number of neurons lost in the brain of patients with AD. Also, currently available drug candidates intervene too late. Stem cells have improved characteristics of self-renewal, proliferation, differentiation, and recombination with the advent of stem cell technology and the transformation of these cells into different types of central nervous system neurons and glial cells. Stem cell treatment has been successful in AD animal models. Recent preclinical studies on stem cell therapy for AD have proved to be promising. Cell replacement therapies, such as human embryonic stem cells or induced pluripotent stem cell-derived neural cells, have the potential to treat patients with AD, and human clinical trials are ongoing in this regard. However, many steps still need to be taken before stem cell therapy becomes a clinically feasible treatment for human AD and related diseases. This paper reviews the pathophysiology of AD and the application prospects of related stem cells based on cell type.
Collapse
Affiliation(s)
- Xin-Yu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
| | - Lin-Po Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
| | - Lan Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China
- Tianjin Key Laboratory of Acupuncture and Moxibustion, Tianjin 300381, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
40
|
Kratzer I, Ek J, Stolp H. The molecular anatomy and functions of the choroid plexus in healthy and diseased brain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183430. [PMID: 32750317 DOI: 10.1016/j.bbamem.2020.183430] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 01/16/2023]
Abstract
The choroid plexus (CP) is located in the ventricular system of the brain (one in each ventricle), and the CP epithelial cells form an important barrier between the blood and the cerebrospinal fluid (CSF). Their main function comprises CSF secretion, maintenance of brain homeostasis, signalling, and forming a neuroprotective barrier against harmful external and internal compounds. The CPs mature early and demonstrate expressional changes of barrier-specific genes and proteins related to location and developmental stage of the CP. Important proteins for the barrier function include tight junction proteins, numerous transporters and enzymes. Natural senescence leads to structural changes in the CP cells and reduced or loss of function, while further loss of CP function and changes in immune status may be relevant in neurodegenerative diseases such as Alzheimer's disease and Multiple Sclerosis. Neuroprotective genes expressed at CPs may be unexplored targets for new therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ingrid Kratzer
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028 CNRS UMR 5292, University Claude Bernard Lyon 1, 69008 Lyon, France; Friedensgasse 3, 8010 Graz, Austria.
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 40530 Göteborg, Sweden.
| | - Helen Stolp
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW0 1TU, UK.
| |
Collapse
|
41
|
Hsu HW, Rodriguez-Ortiz CJ, Lim SL, Zumkehr J, Kilian JG, Vidal J, Kitazawa M. Copper-Induced Upregulation of MicroRNAs Directs the Suppression of Endothelial LRP1 in Alzheimer's Disease Model. Toxicol Sci 2020; 170:144-156. [PMID: 30923833 DOI: 10.1093/toxsci/kfz084] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic exposure to copper and its dyshomeostasis have been linked to accelerated cognitive decline and potentially increasing risk for Alzheimer's disease (AD). We and others have previously demonstrated that exposure to copper through drinking water significantly increased parenchymal amyloid-beta (Aβ) plaques and decreased endothelial low-density lipoprotein receptor-related protein 1 (LRP1) in mouse models of AD. In this study, we determined the underlying mechanisms that microRNA critically mediated the copper-induced loss of endothelial LRP1. In human primary microvascular endothelial cells (MVECs), microRNA-200b-3p, -200c-3p, and -205-5p were significantly elevated within the 24-h exposure to copper and returned to baseline after 48-h postexposure, which corresponded with the temporal change of LRP1 expression in these cells. Transient expression of synthetic microRNA-200b-3p, -200c-3p, or -205-5p on MVECs significantly decreased endothelial LRP1, and cotreatment of synthetic antagomirs effectively prevented the loss of LRP1 during copper exposure, collectively supporting the key regulatory role of these microRNAs in copper-induced loss of LRP1. In mice, a significant reduction of LRP1 in cortical vasculature was evident following 9 months exposure to 1.3 ppm copper in drinking water, although the levels of cortical microRNA-205-5p, -200b-3p, and -200c-3p were only marginally elevated. This, however, correlated with increased vascular accumulation of Aβ and impairment of spatial memory, indicating that copper exposure has the pivotal role in the vascular damage and development of cognitive decline.
Collapse
Affiliation(s)
- Heng-Wei Hsu
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Siok Lam Lim
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Joannee Zumkehr
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Jason G Kilian
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Janielle Vidal
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health (COEH), Department of Medicine, University of California, Irvine, California 92617-1830
| |
Collapse
|
42
|
He Y, Ruganzu JB, Jin H, Peng X, Ji S, Ma Y, Zheng L, Yang W. LRP1 knockdown aggravates Aβ 1-42-stimulated microglial and astrocytic neuroinflammatory responses by modulating TLR4/NF-κB/MAPKs signaling pathways. Exp Cell Res 2020; 394:112166. [PMID: 32645395 DOI: 10.1016/j.yexcr.2020.112166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/02/2020] [Accepted: 07/05/2020] [Indexed: 12/22/2022]
Abstract
Neuroinflammation is an important pathological feature and an early event in the pathogenesis of Alzheimer's disease (AD), which is characterized by activation of microglia and astrocytes. Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic receptor that is abundantly expressed in neurons, microglia, and astrocytes, and plays a critical role in AD pathogenesis. There is increasing evidence to show that LRP1 regulates inflammatory responses by modulating the release of pro-inflammatory cytokines and phagocytosis. However, the effects of LRP1 on β-amyloid protein (Aβ)-induced microglial and astrocytic neuroinflammatory responses and its underlying mechanisms have not been studied in detail. In the present study, knockdown of LRP1 significantly enhanced Aβ1-42-stimulated neuroinflammation by increasing the production of pro-inflammatory cytokines in both BV2 microglial cells and mouse primary astrocytes. Furthermore, it is revealed that LRP1 knockdown further led to the activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways. The phosphorylation of IκBα, p38, and JNK was significantly up-regulated in LRP1 knockdown BV2 microglial cells and primary astrocytes. Meanwhile, LRP1 knockdown increased expression of the NF-κB p65 subunit in the nucleus while decreased its expression in the cytoplasm. Besides, the upstream signaling adaptor molecules such as toll-like receptor 4 (TLR4), myeloid differentiation primary response protein 88 (MyD88), and tumor necrosis factor receptor-associated factor 6 (TRAF6) were also further increased. Moreover, blockade of NF-κB, p38, and JNK inhibited the production of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) induced by the knockdown of LRP1. Taken together, these findings indicated that LRP1 as an effective therapeutic target against AD and other neuroinflammation related diseases.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Yanbing Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Liming Zheng
- Basic Medical Experimental Teaching Center, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
43
|
Barker RM, Holly JMP, Biernacka KM, Allen-Birt SJ, Perks CM. Mini Review: Opposing Pathologies in Cancer and Alzheimer's Disease: Does the PI3K/Akt Pathway Provide Clues? Front Endocrinol (Lausanne) 2020; 11:403. [PMID: 32655497 PMCID: PMC7324530 DOI: 10.3389/fendo.2020.00403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
This minireview is a brief overview examining the roles of insulin-like growth factors (IGFs) and the PI3K/Akt pathway in two apparently unconnected diseases: Alzheimer's dementia and cancer. For both, increased age is a major risk factor, and, in accord with the global rise in average life expectancy, their prevalence is also increasing. Cancer, however, involves excessive cell proliferation and metastasis, whereas Alzheimer's disease (AD) involves cell death and tissue destruction. The apparent "inverse" nature of these disease states is examined here, but also some important commonalities in terms of the PI3K/Akt pathway, glucose utilization and cell deregulation/death. The focus here is on four key molecules associated with this pathway; notably, the insulin receptor substrate 1 (IRS-1), cellular tumor antigen p53 (p53), peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (PIN1) and low-density lipoprotein receptor-related protein-1 (LRP1), all previously identified as potential therapeutic targets for both diseases. The insulin-resistant state, commonly reported in AD brain, results in neuronal glucose deprivation, due to a dampening down of the PI3K/Akt pathway, including overactivity of the mammalian target of rapamycin 1 (mTORC1) complex, hyperphosphorylation of p53 and neuronal death. This contrasts with cancer, where there is overstimulation of the PI3K/Akt pathway and the suppression of mTORC1 and p53, enabling abundant energy and unrestrained cell proliferation. Although these disease states appear to be diametrically opposed, the same key molecules are controlling pathology and, with differential targeting of therapeutics, may yet provide a beneficial outcome for both.
Collapse
Affiliation(s)
- Rachel M. Barker
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Jeff M. P. Holly
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Kalina M. Biernacka
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Shelley J. Allen-Birt
- Molecular Neurobiology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, Southmead Hospital, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
44
|
He Y, Ruganzu JB, Zheng Q, Wu X, Jin H, Peng X, Ding B, Lin C, Ji S, Ma Y, Yang W. Silencing of LRP1 Exacerbates Inflammatory Response Via TLR4/NF-κB/MAPKs Signaling Pathways in APP/PS1 Transgenic Mice. Mol Neurobiol 2020; 57:3727-3743. [PMID: 32572761 DOI: 10.1007/s12035-020-01982-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022]
Abstract
Activation of glial cells (including microglia and astrocytes) appears central to the initiation and progression of neuroinflammation in Alzheimer's disease (AD). The low-density lipoprotein receptor-related protein 1 (LRP1) is a major receptor for amyloid-β (Aβ), which plays a critical role in AD pathogenesis. LRP1 regulates inflammatory response by modulating the release of pro-inflammatory cytokines and phagocytosis. However, the effects of LRP1 on microglia- and astrocytic cell-mediated neuroinflammation and their underlying mechanisms in AD remain unclear. Therefore, using APP/PS1 transgenic mice, we found that LRP1 is downregulated during disease progression. Silencing of brain LRP1 markedly exacerbated AD-related neuropathology including Aβ deposition, neuroinflammation, and synaptic and neuronal loss, which was accompanied by a decline in spatial cognitive ability. Further mechanistic study revealed that silencing of LRP1 initiated neuroinflammation by increasing microgliosis and astrogliosis, enhancing pro-inflammatory cytokine production, and regulating toll-like receptor 4 (TLR4)-mediated activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. Taken together, these findings indicated that LRP1 suppresses microglia and astrocytic cell activation by modulating TLR4/NF-κB/MAPK signaling pathways. Our results further provide insights into the role of LRP1 in AD pathogenesis and highlight LRP1 as a potential therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - Quzhao Zheng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiangyuan Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - Bo Ding
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chengheng Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - Yanbing Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi province, China.
| |
Collapse
|
45
|
Popović N, Morales-Delgado N, Vidal Mena D, Alonso A, Pascual Martínez M, Caballero Bleda M, Popović M. Verapamil and Alzheimer's Disease: Past, Present, and Future. Front Pharmacol 2020; 11:562. [PMID: 32431612 PMCID: PMC7214748 DOI: 10.3389/fphar.2020.00562] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
Verapamil is a phenylalkylamine class calcium channel blocker that for half a century has been used for the treatment of cardiovascular diseases. Nowadays, verapamil is also considered as a drug option for the treatment of several neurological and psychiatric disorders, such as cluster headache, bipolar disorders, epilepsy, and neurodegenerative diseases. Here, we review insights into the potential preventive and therapeutic role of verapamil on Alzheimer’s disease (AD) based on limited experimental and clinical data. Pharmacological studies have shown that verapamil has a wide therapeutic spectrum, including antihypertensive, anti-inflammatory, and antioxidative effects, regulation of the blood-brain barrier function, due to its effect on P-glycoprotein, as well as adjustment of cellular calcium homeostasis, which may result in the delay of AD onset or ameliorate the symptoms of patients. However, the majority of the AD individuals are on polypharmacotherapy, and the interactions between verapamil and other drugs need to be considered. Therefore, for an appropriate and successful AD treatment, a personalized approach is more than necessary. A well-known narrow pharmacological window of verapamil efficacy may hinder this approach. It is therefore important to note that the verapamil efficacy may be conditioned by different factors. The onset, grade, and brain distribution of AD pathological hallmarks, the time-sequential appearances of AD-related cognitive and behavioral dysfunction, the chronobiologic and gender impact on calcium homeostasis and AD pathogenesis may somehow be influencing that success. In the future, such insights will be crucial for testing the validity of verapamil treatment on animal models of AD and clinical approaches.
Collapse
Affiliation(s)
- Natalija Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Nicanor Morales-Delgado
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain.,Department of Histology and Anatomy, Faculty of Medicine, University of Miguel Hernández, Sant Joan Alacant, Spain
| | - David Vidal Mena
- Neurological Unit, University Hospital "Santa Lucia", Cartagena, Spain
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | | | - María Caballero Bleda
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Miroljub Popović
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain.,Institute of Biomedical Research of Murcia (IMIB), Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| |
Collapse
|
46
|
Ferrer I, Andrés-Benito P. White matter alterations in Alzheimer's disease without concomitant pathologies. Neuropathol Appl Neurobiol 2020; 46:654-672. [PMID: 32255227 PMCID: PMC7754505 DOI: 10.1111/nan.12618] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/23/2020] [Indexed: 12/14/2022]
Abstract
Aims Most individuals with AD neuropathological changes have co‐morbidities which have an impact on the integrity of the WM. This study analyses oligodendrocyte and myelin markers in the frontal WM in a series of AD cases without clinical or pathological co‐morbidities. Methods From a consecutive autopsy series, 206 cases had neuropathological changes of AD; among them, only 33 were AD without co‐morbidities. WM alterations were first evaluated in coronal sections of the frontal lobe in every case. Then, RT‐qPCR and immunohistochemistry were carried out in the frontal WM of AD cases without co‐morbidities to analyse the expression of selected oligodendrocyte and myelin markers. Results WM demyelination was more marked in AD with co‐morbidities when compared with AD cases without co‐morbidities. Regarding the later, mRNA expression levels of MBP, PLP1, CNP, MAG, MAL, MOG and MOBP were preserved at stages I–II/0–A when compared with middle‐aged (MA) individuals, but significantly decreased at stages III–IV/0–C. This was accompanied by reduced expression of NG2 and PDGFRA mRNA, reduced numbers of NG2‐, Olig2‐ and HDAC2‐immunoreactive cells and reduced glucose transporter immunoreactivity. Partial recovery of some of these markers occurred at stages V–VI/B–C. Conclusions The present observations demonstrate that co‐morbidities have an impact on WM integrity in the elderly and in AD, and that early alterations in oligodendrocytes and transcription of genes linked to myelin proteins in WM occur in AD cases without co‐morbidities. These are followed by partial recovery attempts at advanced stages. These observations suggest that oligodendrocytopathy is part of AD.
Collapse
Affiliation(s)
- I Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Bellvitge University Hospital, Barcelona, Spain.,Ministry of Economy and Competitiveness, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - P Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Ministry of Economy and Competitiveness, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
47
|
Jia Y, Wang N, Zhang Y, Xue D, Lou H, Liu X. Alteration in the Function and Expression of SLC and ABC Transporters in the Neurovascular Unit in Alzheimer's Disease and the Clinical Significance. Aging Dis 2020; 11:390-404. [PMID: 32257549 PMCID: PMC7069460 DOI: 10.14336/ad.2019.0519] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/19/2019] [Indexed: 12/17/2022] Open
Abstract
The neurovascular unit (NVU) plays an important role in maintaining the function of the central nervous system (CNS). Emerging evidence has indicated that the NVU changes function and molecules at the early stage of Alzheimer’s disease (AD), which initiates multiple pathways of neurodegeneration. Cell types in the NVU have become attractive targets in the interventional treatment of AD. The NVU transportation system contains a variety of proteins involved in compound transport and neurotransmission. Brain transporters can be classified as members of the solute carrier (SLC) and ATP-binding cassette (ABC) families in the NVU. Moreover, the transporters can regulate both endogenous toxins, including amyloid-beta (Aβ) and xenobiotic homeostasis, in the brains of AD patients. Genome-wide association studies (GWAS) have identified some transporter gene variants as susceptibility loci for late-onset AD. Therefore, the present study summarizes changes in blood-brain barrier (BBB) permeability in AD, identifies the location of SLC and ABC transporters in the brain and focuses on major SLC and ABC transporters that contribute to AD pathology.
Collapse
Affiliation(s)
- Yongming Jia
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Na Wang
- 2Department of Pathophysiology, Basic Medical Science College, Qiqihar Medical University, Qiqihar, China
| | - Yingbo Zhang
- 3College of Pathology, Qiqihar Medical University, Qiqihar, China
| | - Di Xue
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Haoming Lou
- 4Department of Medicinal Chemistry and Chemistry of Chinese Materia Medica, School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xuewei Liu
- 1Department of Neuropharmacology, College of Pharmacy, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
48
|
Keep RF, Jones HC, Drewes LR. This was the year that was: brain barriers and brain fluid research in 2019. Fluids Barriers CNS 2020; 17:20. [PMID: 32138786 PMCID: PMC7059280 DOI: 10.1186/s12987-020-00181-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research published in 2019, as well as addressing current controversies and pressing needs. Topics include recent advances related to: the cerebral endothelium and the neurovascular unit; the choroid plexus, arachnoid membrane; cerebrospinal fluid and the glymphatic hypothesis; the impact of disease states on brain barriers and brain fluids; drug delivery to the brain; and translation of preclinical data to the clinic. This editorial also mourns the loss of two important figures in the field, Malcolm B. Segal and Edward G. Stopa.
Collapse
Affiliation(s)
- Richard F. Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | | | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812 USA
| |
Collapse
|
49
|
Yan L, Xie Y, Satyanarayanan SK, Zeng H, Liu Q, Huang M, Ma Y, Wan JB, Yao X, Su KP, Su H. Omega-3 polyunsaturated fatty acids promote brain-to-blood clearance of β-Amyloid in a mouse model with Alzheimer's disease. Brain Behav Immun 2020; 85:35-45. [PMID: 31132459 DOI: 10.1016/j.bbi.2019.05.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/25/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Amyloid-β (Aβ) plaques is one of the typical pathological hallmark of Alzheimer disease (AD). Accumulating evidence suggests that the imbalance between Aβ production and clearance leads to extracellular Aβ accumulation in the brain. It is reported that the blood-brain barrier (BBB) transport plays a predominant role in Aβ clearance from brain to blood. In the present study, we investigated dynamic alterations of BBB transport function in the early disease stage of AD using APPswe/PS1dE9 C57BL/6J (APP/PS1) transgenic mice. Our results showed that the expression of lipoprotein receptor-related protein 1 (LRP-1), a main efflux transporter of BBB, started to decrease at the age of 4 months old. Interestingly, supplementing with fish oil which is rich in omega-3 polyunsaturated fatty acids (PUFAs) significantly enhanced the expression level of LRP-1 and promoted Aβ clearance from the bran to circulation, as revealed by reduced soluble/insoluble Aβ levels and senile plaques in the brain parenchyma and a corresponding increase of Aβ levels in plasma. Besides, fish oil supplement significantly inhibited the NF-κB activation, reduced the expression of interleukin-1β and tumor necrosis factor-α, and suppressed the glial activation in APP/PS1 mice. The results of the study provide evidence that BBB transport function could be impaired at a very early disease stage, which might contribute to Aβ pathological accumulation in AD, and omega-3 PUFAs intervention could be an effective strategy for the prevention of the progression of AD through promoting Aβ clearance from brain-to-blood.
Collapse
Affiliation(s)
- Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Youna Xie
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Haitao Zeng
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Miaodan Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yuemeng Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiaoli Yao
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kuan-Pin Su
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
50
|
5-Caffeoylquinic Acid Ameliorates Cognitive Decline and Reduces Aβ Deposition by Modulating Aβ Clearance Pathways in APP/PS2 Transgenic Mice. Nutrients 2020; 12:nu12020494. [PMID: 32075202 PMCID: PMC7071270 DOI: 10.3390/nu12020494] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/13/2022] Open
Abstract
The accumulation of amyloid β (Aβ) in the brain is a major pathological feature of Alzheimer's disease (AD). In our previous study, we demonstrated that coffee polyphenols (CPP) prevent cognitive dysfunction and Aβ deposition in the brain of an APP/PS2 transgenic mouse AD model. The underlying mechanisms, however, remain to be elucidated. Here, we investigated the effects of the chronic administration of 5-caffeoylquinic acid (5-CQA), the most abundant component of CPP, on cognitive dysfunction in APP/PS2 mice to identify the role of CPP in Aβ elimination. Relative to the untreated controls, the mice fed a 5-CQA-supplemented diet showed significant improvements in their cognitive function assessed by Y-maze and novel object recognition tests. Histochemical analysis revealed that 5-CQA substantially reduced Aβ plaque formation and neuronal loss in the hippocampi. Moreover, 5-CQA upregulated the gene encoding low-density lipoprotein receptor-related protein 1, an Aβ efflux receptor, and normalized the perivascular localization of aquaporin 4, which facilitates Aβ clearance along the paravascular pathway. These results suggest that 5-CQA reduces Aβ deposition in the brain by modulating the Aβ clearance pathways and ameliorating cognitive decline and neuronal loss in APP/PS2 mice. Thus, 5-CQA may be effective in preventing cognitive dysfunction in AD.
Collapse
|