1
|
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
|
2
|
Xia X, Wei Y, Huang Q, Zhou Y, Wang X, Shi Y, Yang X, Yang W, Zhang Y, Lei T, Huang Y, Li H, Qin M, Gao H. Counteracting Alzheimer's disease via normalizing neurovascular unit with a self-regulated multi-functional nano-modulator. Acta Pharm Sin B 2024; 14:5464-5478. [PMID: 39807324 PMCID: PMC11725031 DOI: 10.1016/j.apsb.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 01/16/2025] Open
Abstract
The neurovascular unit (NVU) is highly responsible for cerebral homeostasis and its dysfunction emerges as a critical contributor to Alzheimer's disease (AD) pathology. Hence, rescuing NVU dysfunction might be a viable approach to AD treatments. Here, we fabricated a self-regulated muti-functional nano-modulator (siR/PIO@RP) that can intelligently navigate to damaged blood-brain barrier and release therapeutical cargoes for synergetic AD therapy. The resulting siR/PIO@RP enables self-regulation of its distribution in accordance with the physio/pathological state (low/high RAGE expression) of the target site via a feedback loop. siR/PIO@RP is capable of performing intricate tasks and goes beyond the capabilities of single-target therapeutic agents utilized in AD therapy, such as reducing cerebral Aβ load, relieving neuroinflammation, and alleviating the dysfunction of NVU. Overall, the current study provides proof of concept that normalizing NVU holds promise as a means of alleviating AD symptoms.
Collapse
Affiliation(s)
- Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ya Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qianqian Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yulong Shi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaotong Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenqin Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yiwei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- Key Laboratory of Coarse Cereal Processing, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
3
|
Barrón-González M, Rivera-Antonio AM, Jarillo-Luna RA, Santiago-Quintana JM, Levaro-Loquio D, Pérez-Capistran T, Guerra-Araiza CH, Soriano-Ursúa MA, Farfán-García ED. Borolatonin limits cognitive deficit and neuron loss while increasing proBDNF in ovariectomised rats. Fundam Clin Pharmacol 2024; 38:730-741. [PMID: 38423984 DOI: 10.1111/fcp.12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Borolatonin is a potential therapeutic agent for some neuronal diseases such as Alzheimer's disease (AD). Its administration exerts ameliorative effects such as those induced by the equimolar administration of melatonin in behavioral tests on male rats and in neuronal immunohistochemistry assays. OBJECTIVE In this study, motivated by sex differences in neurobiology and the incidence of AD, the ability of borolatonin to induce changes in female rats was assessed. METHODS Effects of borolatonin were measured by the evaluation of both behavioral and immunohistopathologic approaches; additionally, its ability to limit amyloid toxicity was determined in vitro. RESULTS Surprisingly, behavioral changes were similar to those reported in male rats, but not those evaluated by immunoassays regarding neuronal survival; while pro-brain-derived neurotrophic factor (BDNF) immunoreactivity and the limitation of toxicity by amyloid in vitro were observed for the first time. CONCLUSION Borolatonin administration induced changes in female rats. Differences induced by the administration of borolatonin or melatonin could be related to the differences in the production of steroid hormones in sex dependence. Further studies are required to clarify the possible mechanism and origin of differences in disturbed memory caused by the gonadectomy procedure between male and female rats.
Collapse
Affiliation(s)
- Mónica Barrón-González
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Astrid M Rivera-Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Rosa A Jarillo-Luna
- Laboratorio de Morfología, Sección de Estudios de Posgrado e Investigación, ESM-IPN, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - José M Santiago-Quintana
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - David Levaro-Loquio
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Teresa Pérez-Capistran
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Christian H Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, Mexico
| | - Marvin A Soriano-Ursúa
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| | - Eunice D Farfán-García
- Academias de Fisiología, Bioquímica Médica, y Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Ciudad de México, Mexico
| |
Collapse
|
4
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
5
|
Fan W, Shao K, Luo M. Structural View of Cryo-Electron Microscopy-Determined ATP-Binding Cassette Transporters in Human Multidrug Resistance. Biomolecules 2024; 14:231. [PMID: 38397468 PMCID: PMC10886794 DOI: 10.3390/biom14020231] [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: 12/19/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
ATP-binding cassette (ABC) transporters, acting as cellular "pumps," facilitate solute translocation through membranes via ATP hydrolysis. Their overexpression is closely tied to multidrug resistance (MDR), a major obstacle in chemotherapy and neurological disorder treatment, hampering drug accumulation and delivery. Extensive research has delved into the intricate interplay between ABC transporter structure, function, and potential inhibition for MDR reversal. Cryo-electron microscopy has been instrumental in unveiling structural details of various MDR-causing ABC transporters, encompassing ABCB1, ABCC1, and ABCG2, as well as the recently revealed ABCC3 and ABCC4 structures. The newly obtained structural insight has deepened our understanding of substrate and drug binding, translocation mechanisms, and inhibitor interactions. Given the growing body of structural information available for human MDR transporters and their associated mechanisms, we believe it is timely to compile a comprehensive review of these transporters and compare their functional mechanisms in the context of multidrug resistance. Therefore, this review primarily focuses on the structural aspects of clinically significant human ABC transporters linked to MDR, with the aim of providing valuable insights to enhance the effectiveness of MDR reversal strategies in clinical therapies.
Collapse
Affiliation(s)
| | | | - Min Luo
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore; (W.F.); (K.S.)
| |
Collapse
|
6
|
Thorwald MA, Silva J, Head E, Finch CE. Amyloid futures in the expanding pathology of brain aging and dementia. Alzheimers Dement 2023; 19:2605-2617. [PMID: 36536382 PMCID: PMC10271937 DOI: 10.1002/alz.12896] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/06/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022]
Abstract
Positron emission tomography (PET) imaging studies of Alzheimer's disease (AD) patients show progressive increases of fibrillar Aβ-amyloid. Because current PET ligands underestimate nonfibrillar forms, we assayed soluble Aβ in AD and controls. To identify the mechanisms responsible for soluble Aβ in AD brains, we examined lipid rafts (LRs), where amyloid precursor protein (APP) is enzymatically processed. Frontal cortex was compared with cerebellum, which has minimal AD pathology. Compared with cognitively normal controls (CTL; Braak 0-1), elevations of soluble Aβ40 and Aβ42 were similar for intermediate- and later-stage AD (Braak 2-3 and 4-6). Clinical-grade AD showed a greater increase in soluble Aβ40 than Aβ42 relative to CTL. LR raft yield per gram AD frontal cortex was 20% below that of controls, whereas cerebellar LR did not differ by Braak score. The extensive overlap of soluble Aβ levels in controls with AD contrasts with the PET findings on fibrillar Aβ. These findings further support fibrillar Aβ as a biomarker for AD treatments and show the need for more detailed postmortem analysis of diverse soluble and insoluble Aβ aggregates in relation to PET.
Collapse
Affiliation(s)
- Max A. Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
| | - Justine Silva
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Elizabeth Head
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Caleb E. Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA
- Dornsife College, University of Southern California, Los Angeles, CA
| |
Collapse
|
7
|
Tekin E, Aslan Karakelle N, Dinçer S. Effects of taurine on metal cations, transthyretin and LRP-1 in a rat model of Alzheimer's disease. J Trace Elem Med Biol 2023; 79:127219. [PMID: 37229981 DOI: 10.1016/j.jtemb.2023.127219] [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: 01/08/2023] [Revised: 04/28/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Researches on diagnosis and treatment of Alzheimer's disease, the most common type of dementia, are still ongoing. Taurine is frequently used in Alzheimer's disease models due to its protective effects. Metal cation dyshomeostasis is an important etiological factor for Alzheimer's disease. Transthyretin protein is thought to act as a transporter for the Aβ protein that accumulates in the brain and is eliminated in the liver and kidneys via the LRP-1 receptor. However, the effect of taurine on this mechanisms is not fully known. METHODS 30 male rats, aged 28 ± 4 months, were divided into 5 groups (n = 6) as follows: control group, sham group, Aβ 1-42 group, taurine group and taurine+Aβ 1-42 group. Oral taurine pre-supplementation was given as 1000 mg/kg-body weight/day for 6 weeks to taurine and taurine+Aβ 1-42 groups. RESULTS Plasma copper, heart transthyretin and Aβ 1-42, brain and kidney LRP-1 levels were found to be decreased in the Aβ 1-42 group. Brain transthyretin was higher in taurine+Aβ 1-42 group and brain Aβ 1-42 was higher in Aβ 1-42 and taurine+Aβ 1-42 groups. CONCLUSION Taurine pre-supplementation maintained cardiac transthyretin levels, decreased cardiac Aβ 1-42 levels and increased brain and kidney LRP-1 levels. Taurine may have a potential to be used as a protective agent for aged people at high risk for Alzheimer's disease.
Collapse
Affiliation(s)
- Esra Tekin
- Gazi University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| | - Nida Aslan Karakelle
- Lokman Hekim University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| | - Sibel Dinçer
- Gazi University Faculty of Medicine, Department of Physiology, Ankara, Turkey.
| |
Collapse
|
8
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
9
|
Barrón-González M, Rosales-Hernández MC, Abad-García A, Ocampo-Néstor AL, Santiago-Quintana JM, Pérez-Capistran T, Trujillo-Ferrara JG, Padilla-Martínez II, Farfán-García ED, Soriano-Ursúa MA. Synthesis, In Silico, and Biological Evaluation of a Borinic Tryptophan-Derivative That Induces Melatonin-like Amelioration of Cognitive Deficit in Male Rat. Int J Mol Sci 2022; 23:3229. [PMID: 35328650 PMCID: PMC8952423 DOI: 10.3390/ijms23063229] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
Preclinical and clinical evidence supports melatonin and its analogues as potential treatment for diseases involving cognitive deficit such as Alzheimer's disease. In this work, we evaluated by in silico studies a set of boron-containing melatonin analogues on MT1 and MT2 receptors. Then, we synthesized a compound (borolatonin) identified as potent agonist. After chemical characterization, its evaluation in a rat model with cognitive deficit showed that it induced ameliorative effects such as those induced by equimolar administration of melatonin in behavioral tests and in neuronal immunohistochemistry assays. Our results suggest the observed effects are by means of action on the melatonin system. Further studies are required to clarify the mechanism(s) of action, as the beneficial effects on disturbed memory by gonadectomy in male rats are attractive.
Collapse
Affiliation(s)
- Mónica Barrón-González
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| | - Martha C. Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico;
| | - Antonio Abad-García
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| | - Ana L. Ocampo-Néstor
- Departamento de Nefrología, Hospital General de México, “Dr. Eduardo Liceaga”, Dr. Balmis 148, Alc. Cuauhtémoc, Mexico City 06720, Mexico;
| | - José M. Santiago-Quintana
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional, Av. Acueducto s/n Barrio la Laguna, Ticomán, Mexico City 07340, Mexico;
| | - Teresa Pérez-Capistran
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| | - José G. Trujillo-Ferrara
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| | - Itzia I. Padilla-Martínez
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología del Instituto Politécnico Nacional, Av. Acueducto s/n Barrio la Laguna, Ticomán, Mexico City 07340, Mexico;
| | - Eunice D. Farfán-García
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| | - Marvin A. Soriano-Ursúa
- Academias de Fisiología, Bioquímica Médica, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, Col. Casco de Santo Tomás, Alc. Miguel Hidalgo, Mexico City 11340, Mexico; (M.B.-G.); (A.A.-G.); (J.M.S.-Q.); (T.P.-C.); (J.G.T.-F.)
| |
Collapse
|
10
|
Hasegawa M, Kitaguchi N, Takechi H, Kawaguchi K, Ito K, Kato T, Kato M, Nii N, Yamada S, Ohashi A, Koide S, Hayashi H, Takahashi K, Inaguma D, Yuzawa Y, Tsuboi N. First-in-human study of blood amyloid β removal from early Alzheimer's disease patients with normal kidney function. Ther Apher Dial 2022; 26:529-536. [PMID: 35294796 DOI: 10.1111/1744-9987.13827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/24/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Amyloid β (Aβ) is a brain protein that causes Alzheimer's disease (AD). This study aimed to verify whether hemadsorption using a hexadecyl-alkylated cellulose bead (HexDC) column removes blood Aβ and brain Aβ accumulation in mild cognitive impairment/mild AD cases with normal kidney function. METHODS Two patients with positive Aβ on brain imaging underwent HexDC hemadsorption weekly for 6 months. RESULTS The Aβ removal efficiency of HexDC was 87-99%. Aβ1-40 /Aβ1-42 influx into the blood in one session was 596/56 ng and 489/48 ng for Case A and Case B, respectively. Although brain Aβ accumulation did not clearly change after six months of hemadsorption, cognitive functions measured by the two tests were maintained or slightly improved. CONCLUSION Blood Aβ removal was performed in two early AD patients with normal kidney function without adverse events, and it slightly improved or maintained cognitive function. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Midori Hasegawa
- Department of Nephrology, Fujita Health University School of Medicine
| | - Nobuya Kitaguchi
- Faculty of Clinical Engineering, Fujita Health University School of Health Sciences
| | - Hajime Takechi
- Department of Geriatrics and Cognitive Disorders, Fujita Health University School of Medicine
| | - Kazunori Kawaguchi
- Faculty of Clinical Engineering, Fujita Health University School of Health Sciences
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology
| | - Takashi Kato
- Department of Clinical and Experimental Neuroimaging, National Center for Geriatrics and Gerontology
| | - Masao Kato
- Center of Blood Purification, Fujita Health University Hospital
| | - Norio Nii
- Center of Blood Purification, Fujita Health University Hospital
| | - Sachie Yamada
- Center of Blood Purification, Fujita Health University Hospital
| | - Atsushi Ohashi
- Faculty of Clinical Engineering, Fujita Health University School of Health Sciences
| | - Shigehisa Koide
- Department of Nephrology, Fujita Health University School of Medicine
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine
| | - Kazuo Takahashi
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine
| | - Daijo Inaguma
- Department of Internal Medicine, Fujita Health University Bantane Hospital
| | - Yukio Yuzawa
- Department of Nephrology, Fujita Health University School of Medicine
| | - Naotake Tsuboi
- Department of Nephrology, Fujita Health University School of Medicine
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Monllor P, Giraldo E, Badia MC, de la Asuncion JG, Alonso MD, Lloret A, Vina J. Serum Levels of Clusterin, PKR, and RAGE Correlate with Amyloid Burden in Alzheimer's Disease. J Alzheimers Dis 2021; 80:1067-1077. [PMID: 33646167 DOI: 10.3233/jad-201443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia and biomarkers are essential to help in the diagnosis of this disease. Image techniques and cerebrospinal fluid (CSF) biomarkers are limited in their use because they are expensive or invasive. Thus, the search for blood-borne biomarkers is becoming central to the medical community. OBJECTIVE The main objective of this study is the evaluation of three serum proteins as potential biomarkers in AD patients. METHODS We recruited 27 healthy controls, 19 mild cognitive impairment patients, and 17 AD patients. Using the recent A/T/N classification we split our population into two groups (AD and control). We used ELISA kits to determine Aβ42, tau, and p-tau in CSF and clusterin, PKR, and RAGE in serum. RESULTS The levels of serum clusterin, PKR, and RAGE were statistically different in the AD group compared to controls. These proteins showed a statistically significant correlation with CSF Aβ42. So, they were selected to generate an AD detection model showing an AUC-ROC of 0.971 (CI 95%, 0.931-0.998). CONCLUSION The developed model based on serum biomarkers and other co-variates could reflect the AD core pathology. So far, not one single blood-biomarker has been described, with effectiveness offering high sensitivity and specificity. We propose that the complexity of AD pathology could be reflected in a set of biomarkers also including clinical features of the patients.
Collapse
Affiliation(s)
- Paloma Monllor
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Esther Giraldo
- Department of Biotechnology, Universitat Politècnica de València, Valencia, Spain.,Principe Felipe Research Center, Valencia, Spain
| | | | | | | | - Ana Lloret
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| | - Jose Vina
- Freshage Research Group, Department of Physiology, University of Valencia, CIBERFES-ISCIII, INCLIVA, Valencia, Spain
| |
Collapse
|
16
|
Shin SJ, Nam Y, Park YH, Kim MJ, Lee E, Jeon SG, Bae BS, Seo J, Shim SL, Kim JS, Han CK, Kim S, Lee YY, Moon M. Therapeutic effects of non-saponin fraction with rich polysaccharide from Korean red ginseng on aging and Alzheimer's disease. Free Radic Biol Med 2021; 164:233-248. [PMID: 33422674 DOI: 10.1016/j.freeradbiomed.2020.12.454] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022]
Abstract
Biological aging provokes morbidity and several functional declines, causing older adults more susceptible to a variety of diseases than younger adults. In particular, aging is a major risk factor contributing to non-communicable diseases, such as neurodegenerative disorders. Alzheimer's disease (AD) is an aging-related neurodegenerative disease that is characterized by cognitive deficits and the formation of amyloid plaques formed by the accumulation of amyloid-β (Aβ) peptides. Non-saponin fraction with rich polysaccharide (NFP) from red ginseng, the largest fraction of the components of red ginseng, perform many biological activities. However, it has not been clarified whether the NFP from Korean red ginseng (KRG) has beneficial effects in the aging and AD. First, proteomics analysis was performed in aged brain to identify the effect of NFP on protein changes, and we confirmed that NFP induced changes in proteins related to the neuroprotective- and neurogenic-effects. Next, we investigated (1) the effects of NFP on AD pathologies, such as Aβ deposition, neuroinflammation, neurodegeneration, mitochondrial dysfunction, and impaired adult hippocampal neurogenesis (AHN), in 5XFAD transgenic mouse model of AD using immunostaining; (2) the effect of NFP on Aβ-mediated mitochondrial respiration deficiency in HT22 mouse hippocampal neuronal cells (HT22) using Seahorse XFp analysis; (3) the effect of NFP on cell proliferation using WST-1 analysis; and (4) the effect of NFP on Aβ-induced cognitive dysfunction in 5XFAD mouse model of AD using Y-maze test. Histological analysis indicated that NFP significantly alleviated the accumulation of Aβ, neuroinflammation, neuronal loss, and mitochondrial dysfunction in the subiculum of 5XFAD mouse model of AD. In addition, NFP treatment ameliorated mitochondrial deficits in Aβ-treated HT22 cells. Moreover, NFP treatment significantly increased the AHN and neuritogenesis of neural stem cells in both healthy and AD brains. Furthermore, NFP significantly increased cell proliferation in the HT22 cells. Finally, NFP administration significantly enhanced and restored the cognitive function of healthy and AD mice, respectively. Taken together, NFP treatment demonstrated changes in proteins involved in central nervous system organization/maintenance in aged brain and ameliorates AD pathology. Collectively, our findings suggest that NFP from KRG could be a potential therapeutic candidate for aging and AD treatments.
Collapse
Affiliation(s)
- Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Min-Jeong Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Eunbeen Lee
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Seong Gak Jeon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, 41068, Republic of Korea
| | - Bong-Seok Bae
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro 30, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea
| | - Jiho Seo
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro 30, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea
| | - Sung-Lye Shim
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro 30, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea
| | - Jong-Seok Kim
- Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Chang-Kyun Han
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro 30, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea; Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea.
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Korea Ginseng Corporation, Gajeong-ro 30, Shinseong-dong, Yuseong-gu, Daejeon, 34128, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea; Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
17
|
Kitaguchi N, Kawaguchi K, Sakata M, Aoki H, Yamazaki K, Kaneko M, Kinomura J, Kato M, Hasegawa M, Suzuki N, Mizuno M, Yuzawa Y. Aβ Influx into the Blood Evoked by Different Blood Aβ Removal Systems: A Potential Therapy for Alzheimer's Disease. Neuropsychiatr Dis Treat 2021; 17:2291-2308. [PMID: 34285489 PMCID: PMC8286129 DOI: 10.2147/ndt.s317104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Amyloid-β (Aβ) is a brain protein that causes Alzheimer's disease. We have revealed that extracorporeal blood Aβ-removal systems evoked a large Aβ influx into the blood. This study investigated the system that is more effective in evoking Aβ influx. METHODS Aβ removal activities were compared between hexadecyl-alkylated cellulose beads (HexDC) and fragments of polysulfone hollow fibers (PSf-HFs) in mini-columns to eliminate the filtration effect. Then, adsorptive filtration systems were adapted for PSf hemodialyzers to enhance Aβ adsorption on micropores in the wall of hollow fibers. Plasma Aβ concentrations of patients with renal failure were analyzed during treatment with PSf hemodialyzers alone for 8 h or tandemly connected HexDC and PSf hemodialyzers for 4 h. RESULTS In the in vitro study, Aβ removal efficiency for HexDC was approximately 100% during the 60 min treatment, whereas the removal efficiency for PSf-HF fragments gradually decreased. However, PSf hemodialyzer in adsorptive filtration systems removed Aβs comparably or more than HexDC. Aβ influx into the blood increases time-dependently. Concomitant use of HexDC and PSf hemodialyzer evoked a larger Aβ1-40 influx than that of PSf hemodialyzer alone. However, Aβ1-42 influx by PSf hemodialyzer alone was similar to or a little larger than influx by the combined system. Both systems evoked almost doubled Aβ influx than estimated Aβs existing in the normal brain during the 4 h treatment. CONCLUSION PSf hemodialyzer alone for a longer period and concomitant use of HexDC and PSf hemodialyzer for a shorter time effectively evoked a larger Aβ influx. To evoke Aβ1-42 influx, PSf hemodialyzer alone was effective enough. These findings of devices and treatment time may lead to optimal clinical settings for therapy and prevention of Alzheimer's disease.
Collapse
Affiliation(s)
- Nobuya Kitaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazunori Kawaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Miwa Sakata
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Hiroki Aoki
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Kazunori Yamazaki
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Megumi Kaneko
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Jun Kinomura
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Masao Kato
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Midori Hasegawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | | | | | - Yukio Yuzawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
18
|
Zhang Q, Song Q, Gu X, Zheng M, Wang A, Jiang G, Huang M, Chen H, Qiu Y, Bo B, Tong S, Shao R, Li B, Wang G, Wang H, Hu Y, Chen H, Gao X. Multifunctional Nanostructure RAP-RL Rescues Alzheimer's Cognitive Deficits through Remodeling the Neurovascular Unit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001918. [PMID: 33511002 PMCID: PMC7816710 DOI: 10.1002/advs.202001918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/02/2020] [Indexed: 05/21/2023]
Abstract
Cerebrovascular dysfunction characterized by the neurovascular unit (NVU) impairment contributes to the pathogenesis of Alzheimer's disease (AD). In this study, a cerebrovascular-targeting multifunctional lipoprotein-biomimetic nanostructure (RAP-RL) constituted with an antagonist peptide (RAP) of receptor for advanced glycation end-products (RAGE), monosialotetrahexosyl ganglioside, and apolipoprotein E3 is developed to recover the functional NVU and normalize the cerebral vasculature. RAP-RL accumulates along the cerebral microvasculature through the specific binding of RAP to RAGE, which is overexpressed on cerebral endothelial cells in AD. It effectively accelerates the clearance of perivascular Aβ, normalizes the morphology and functions of cerebrovasculature, and restores the structural integrity and functions of NVU. RAP-RL markedly rescues the spatial learning and memory in APP/PS1 mice. Collectively, this study demonstrates the potential of the multifunctional nanostructure RAP-RL as a disease-modifying modality for AD treatment and provides the proof of concept that remodeling the functional NVU may represent a promising therapeutic approach toward effective intervention of AD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Qingxiang Song
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Xiao Gu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Mengna Zheng
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Antian Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Gan Jiang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Meng Huang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Huan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yu Qiu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Bin Bo
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Shanbao Tong
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Rong Shao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Binyin Li
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Gang Wang
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Hao Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yongbo Hu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
- Institute of Interdisciplinary Integrative Biomedical ResearchShuguang HospitalShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| |
Collapse
|
19
|
Kirov II, Sollberger M, Davitz MS, Glodzik L, Soher BJ, Babb JS, Monsch AU, Gass A, Gonen O. Global brain volume and N-acetyl-aspartate decline over seven decades of normal aging. Neurobiol Aging 2020; 98:42-51. [PMID: 33232854 DOI: 10.1016/j.neurobiolaging.2020.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/07/2020] [Accepted: 10/24/2020] [Indexed: 10/23/2022]
Abstract
We characterize the whole-brain N-acetyl-aspartate (WBNAA) and brain tissue fractions across the adult lifespan and test the hypothesis that, despite age-related atrophy, neuronal integrity (reflected by WBNAA) is preserved in normal aging. Two-hundred-and-seven participants: 133 cognitively intact older adults (73.6 ± 7.4 mean ± standard deviation, range: 60-90 year old) and 84 young (37.9 ± 11, range: 21-59 year old) were scanned with proton magnetic resonance spectroscopy and T1-weighted MRI. Their WBNAA, fractional brain parenchyma, and gray and white matter volumes (fBPV, fGM, and fWM) were compared and modeled as functions of age and sex. Compared with young, older-adults' WBNAA was lower by ~35%, and fBPV, fGM and fWM were lower by ~10%. Linear regressions found 0.5%/year WBNAA and 0.2%/year fBPV and fGM declines, whereas fWM rose to age ~40 years, and declined thereafter. fBPV and fGM were 1.8% and 4% higher in women, with no sex decline rates difference. We conclude that contrary to our hypothesis, atrophy was accompanied by WBNAA decline. Across the entire age range, women's brains showed less atrophy than men's. Formulas to estimate WBNAA and brain tissue fractions in healthy adults are provided to help differentiate normal from abnormal aging.
Collapse
Affiliation(s)
- Ivan I Kirov
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Marc Sollberger
- University Department of Geriatric Medicine FELIX PLATTER, Memory Clinic, Basel, Switzerland; Department of Neurology, University Hospital, Basel, Switzerland
| | - Matthew S Davitz
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Brian J Soher
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - James S Babb
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Andreas U Monsch
- University Department of Geriatric Medicine FELIX PLATTER, Memory Clinic, Basel, Switzerland
| | - Achim Gass
- Department of Neurology/Neuroimaging, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Oded Gonen
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
20
|
Zhao Y, Zeng C, Li X, Yang T, Kuang X, Du J. Klotho overexpression improves amyloid-β clearance and cognition in the APP/PS1 mouse model of Alzheimer's disease. Aging Cell 2020; 19:e13239. [PMID: 32964663 PMCID: PMC7576297 DOI: 10.1111/acel.13239] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia, characterized by the presence of amyloid-β (Aβ) plaques. We previously reported that Klotho lowered Aβ levels in the brain and protected against cognitive deficits in amyloid precursor protein/presenilin 1(APP/PS1) mice. However, the underlying mechanism remains unclear. In this study, we induced intracerebral Klotho overexpression in 13-month-old APP/PS1 mice by injecting lentivirus that carried full-length mouse Klotho cDNA in the lateral ventricle of the brain. We examined the effects of Klotho overexpression on cognition, Aβ burden, Aβ-related neuropathology, microglia transformation, and Aβ transport systems in vivo. Additionally, we investigated the effects of Klotho on Aβ transport at the blood-cerebrospinal fluid barrier by knocking down Klotho in primary human choroid plexus epithelial cells (HCPEpiCs). The upregulation of Klotho levels in the brain and serum significantly ameliorated Aβ burden, neuronal and synaptic loss and cognitive deficits in aged APP/PS1 mice. Klotho treatment significantly inhibited NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) and the subsequent transformation of microglia to the M2 type that may enhance microglia-mediated Aβ clearance. Meanwhile, Klotho overexpression also regulated Aβ transporter expression, which may promote Aβ transporter-mediated Aβ clearance. Moreover, the ability of HCPEpiCs to transport Aβ in vitro was also significantly impaired by Klotho knockdown. Given the neuroprotective effect of Klotho overexpression, the present findings suggest that Klotho should be further investigated as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Chen‐Ye Zeng
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Xiao‐Hong Li
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Ting‐Ting Yang
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| | - Jun‐Rong Du
- Department of Pharmacology, Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of PharmacySichuan UniversityChengduChina
| |
Collapse
|
21
|
Kitaguchi N, Tatebe H, Sakai K, Kawaguchi K, Matsunaga S, Kitajima T, Tomizawa H, Kato M, Sugiyama S, Suzuki N, Mizuno M, Takechi H, Nakai S, Hiki Y, Kushimoto H, Hasegawa M, Yuzawa Y, Tokuda T. Influx of Tau and Amyloid-β Proteins into the Blood During Hemodialysis as a Therapeutic Extracorporeal Blood Amyloid-β Removal System for Alzheimer's Disease. J Alzheimers Dis 2020; 69:687-707. [PMID: 31156161 DOI: 10.3233/jad-190087] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The accumulation of amyloid-β protein (Aβ) and tau in the brain is a major pathological change related to Alzheimer's disease. We have continued to develop Extracorporeal Blood Aβ Removal Systems (E-BARS) as a method for enhancing Aβ clearance from the brain. Our previous report revealed that dialyzers effectively remove blood Aβ and evoke large Aβ influxes into the blood, resulting in a decrease in brain Aβ accumulation after initiating hemodialysis, and that patients who underwent hemodialysis had lower brain Aβ accumulation than those who did not. Here, plasma total tau concentrations from 30 patients undergoing hemodialysis were measured using an ultrasensitive immunoassay and compared to those from 11 age-matched controls. Plasma total tau concentrations were higher in patients with renal failure regardless of whether they underwent hemodialysis, suggesting the involvement of the kidneys in tau degradation and excretion. Hemodialyzers effectively removed blood Aβ but not extracorporeal blood tau. The influx of tau into the blood was observed at around the 1 h period during hemodialysis sessions. However, the influx amount of tau was far smaller than that of Aβ. Furthermore, histopathological analysis revealed similar, not significantly less, cerebral cortex phosphorylated tau accumulation between the 17 patients who underwent hemodialysis and the 16 age-matched subjects who did not, although both groups showed sparse accumulation. These findings suggest that hemodialysis may induce both tau and Aβ migration into the blood. However, as a therapeutic strategy for Alzheimer's disease, it may only be effective for removing Aβ from the brain.
Collapse
Affiliation(s)
- Nobuya Kitaguchi
- Faculty of Clinical Engineering, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Harutsugu Tatebe
- Department of Zaitaku (Homecare) Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuyoshi Sakai
- Faculty of Clinical Engineering, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Kazunori Kawaguchi
- Faculty of Clinical Engineering, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Shinji Matsunaga
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoko Kitajima
- Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Masao Kato
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Satoshi Sugiyama
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | | | | | - Hajime Takechi
- Department of Geriatrics and Cognitive Disorders, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Shigeru Nakai
- Faculty of Clinical Engineering, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiyuki Hiki
- Faculty of Clinical Engineering, School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Midori Hasegawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Yukio Yuzawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Takahiko Tokuda
- Department of Molecular Pathobiology of Brain Diseases, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
22
|
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: 16] [Impact Index Per Article: 3.2] [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
|
23
|
Min LJ, Iwanami J, Shudou M, Bai HY, Shan BS, Higaki A, Mogi M, Horiuchi M. Deterioration of cognitive function after transient cerebral ischemia with amyloid-β infusion-possible amelioration of cognitive function by AT 2 receptor activation. J Neuroinflammation 2020; 17:106. [PMID: 32264971 PMCID: PMC7140348 DOI: 10.1186/s12974-020-01775-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Background To promote understanding of the pathogenesis of cognitive impairment or dementia, we explored the potential interaction between transient cerebral ischemia and amyloid-β (Aβ) infusion in mediating cognitive decline and examined the possible ameliorative effect of angiotensin II type 2 (AT2) receptor activation in vascular smooth muscle cells (VSMC) on this cognitive deficit. Methods Adult male wild-type mice (WT) and mice with VSMC-specific AT2 receptor overexpression (smAT2) were subjected to intracerebroventricular (ICV) injection of Aβ1-40. Transient cerebral ischemia was induced by 15 min of bilateral common carotid artery occlusion (BCCAO) 24 h after Aβ injection. Results Aβ injection in WT induced a cognitive decline, whereas BCCAO did not cause a significant cognitive deficit. In contrast, WT with BCCAO following Aβ injection exhibited more marked cognitive decline compared to Aβ injection alone, in concert with increases in superoxide anion production, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and expression of p22phox, p40phox, monocyte chemoattractant protein (MCP)-1 and interleukin (IL)-1β in the hippocampus, and upregulation of RAGE (receptor for advanced glycation end product), an Aβ transporter. BCCAO following Aβ injection further enhanced neuronal pyknosis in the hippocampus, compared with BCCAO or Aβ injection alone. In contrast, smAT2 did not show a cognitive decline, increase in oxidative stress, inflammation, and RAGE level or neuronal pyknosis, which were induced by BCCAO with/without Aβ injection in WT. Conclusions Transient cerebral ischemia might worsen Aβ infusion-mediated cognitive decline and vice versa, with possible involvement of amplified oxidative stress and inflammation and impairment of the RAGE-mediated Aβ clearance system, contributing to exaggerated neuronal degeneration. AT2 receptor activation in VSMC could play an inhibitory role in this cognitive deficit.
Collapse
Affiliation(s)
- Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan.
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masachika Shudou
- Division of Analytical Bio-Medicine, Advanced Research Support Center (ADRES), Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, 791-0295, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan
| |
Collapse
|
24
|
López-González I, Tebé Cordomí C, Ferrer I. Regional Gene Expression of Inflammation and Oxidative Stress Responses Does Not Predict Neurodegeneration in Aging. J Neuropathol Exp Neurol 2020; 76:135-150. [PMID: 28158670 DOI: 10.1093/jnen/nlw117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Brain aging is accompanied by increased oxidative stress and what has been termed "neuroinflammation," which might contribute to age-related neurodegenerative diseases. We analyzed expression in the transcription of innate inflammatory response genes in eleven representative regions including frontal, parietal, inferior temporal, cingulate, occipital, entorhinal cortex, caudate, putamen, thalamus, substantia nigra, and cerebellar vermis in aging human brains. We probed members of the complement system, colony stimulating factor receptors, toll-like receptors, and pro- and anti-inflammatory cytokines in the brains of subjects with no neurological disease and neurofibrillary tangles (mean age: 47.1 ± 5.7 years) and those with no neurological disease and neurofibrillary pathology stages I-II (mean age: 70.6 ± 6.3 years). Although the entorhinal and frontal cortex were most altered, gene regulation patterns did not match regions with increased vulnerability. Analysis of false discovery rate thresholds revealed no differences for any gene in any region between the 2 groups, including cases in which individual comparisons analyzed using Student t or nonparametric tests showed apparent differences between groups. Moreover, gene expression of major anti-oxidative stress responses did not match neuroinflammation in aging or increased regional susceptibility to major neurodegenerative diseases.
Collapse
Affiliation(s)
- Irene López-González
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Bellvitge University Hospital (IL-G, IF); Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat (IL-G, CTC, IF); Faculty of Medicine and Health Sciences, University Rovira i Virgili University, Reus (CTC); Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat (IF); Institute of Neurosciences, University of Barcelona (IF); and CIBERNED (Biomedical Research Center Network for the Study of Neurodegenerative Diseases, Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain (IF)
| | - Cristian Tebé Cordomí
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Bellvitge University Hospital (IL-G, IF); Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat (IL-G, CTC, IF); Faculty of Medicine and Health Sciences, University Rovira i Virgili University, Reus (CTC); Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat (IF); Institute of Neurosciences, University of Barcelona (IF); and CIBERNED (Biomedical Research Center Network for the Study of Neurodegenerative Diseases, Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain (IF)
| | - Isidro Ferrer
- From the Institute of Neuropathology, Service of Pathologic Anatomy, Bellvitge University Hospital (IL-G, IF); Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat (IL-G, CTC, IF); Faculty of Medicine and Health Sciences, University Rovira i Virgili University, Reus (CTC); Department of Pathology and Experimental Therapeutics, University of Barcelona, L'Hospitalet de Llobregat (IF); Institute of Neurosciences, University of Barcelona (IF); and CIBERNED (Biomedical Research Center Network for the Study of Neurodegenerative Diseases, Institute Carlos III, Spanish Ministry of Science and Innovation, Madrid, Spain (IF)
| |
Collapse
|
25
|
Saito Y, Sakata M, Kobayakawa M, Kawachi H, Kawaguchi K, Hiki Y, Kato M, Mori M, Hasegawa M, Ohashi N, Yuzawa Y, Kitaguchi N. Removal of Aβ Oligomers from the Blood: A Potential Therapeutic System for Alzheimer's Disease. Neuropsychiatr Dis Treat 2020; 16:607-627. [PMID: 32210564 PMCID: PMC7064818 DOI: 10.2147/ndt.s241074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Amyloid-β protein (Aβ) is one of the causative proteins of Alzheimer's disease. We have been developing extracorporeal blood Aβ-removal systems as a method for enhancing Aβ clearance from the brain. We reported previously that medical adsorbents and hemodialyzers removed Aβ monomers from peripheral blood, which was associated with influx of Aβ monomers from the brain into the bloodstream. Our intent here was to develop a method to promote clearance of Aβ oligomers and to provide an estimate of the molecular size of intact Aβ oligomers in plasma. METHODS Two hollow-fiber devices with different pore sizes (Membranes A and B) were evaluated as removers of Aβ oligomers with human plasma in vitro. The concomitant removal of Aβ oligomers and monomers was investigated by using Membrane B and hexadecyl alkylated cellulose beads or polysulfone hemodialyzers. Double-filtration plasmapheresis with Membrane A was investigated as an approach for the removal of plasma Aβ oligomers in humans. RESULTS Aβ oligomers were effectively removed by both Membranes A and B. The increase of Aβ oligomers in plasma was observed just after the removal of plasma Aβ oligomers in humans. The intact molecular size of major Aβ oligomers in the plasma was estimated to be larger than albumin at approximately 60 kDa or more. Additionally, the concomitant removal of Aβ monomers and oligomers evoked dissociation of larger Aβ oligomers into smaller ones and monomers. CONCLUSION Aβ oligomers were cleared from plasma both in vitro and in human subjects by using hollow-fiber membranes with large pores, indicating that their intact sizes were mostly larger than 60 kDa. Aβ oligomers in peripheral circulation were increased after some clearances in human. Further investigation will determine whether the Aβ oligomers detected in circulation after clearance were via influx from the brain.
Collapse
Affiliation(s)
- Yuta Saito
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.,Graduate School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan.,Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Miwa Sakata
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Moe Kobayakawa
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Kazunori Kawaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiyuki Hiki
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Masao Kato
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Mayuko Mori
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.,Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Midori Hasegawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Yukio Yuzawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Nobuya Kitaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
26
|
Kant S, Stopa EG, Johanson CE, Baird A, Silverberg GD. Choroid plexus genes for CSF production and brain homeostasis are altered in Alzheimer's disease. Fluids Barriers CNS 2018; 15:34. [PMID: 30541599 PMCID: PMC6291926 DOI: 10.1186/s12987-018-0120-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/03/2018] [Indexed: 11/10/2022] Open
Abstract
Background The roles of the choroid plexus (CP) and cerebrospinal fluid (CSF) production have drawn increasing attention in Alzheimer’s disease (AD) research. Specifically, studies document markedly decreased CSF production and turnover in moderate-to-severe AD. Moreover, reduced CP function and CSF turnover lead to impaired clearance of toxic metabolites, likely promote neuroinflammation, and may facilitate neuronal death during AD progression. We analyzed CP gene expression in AD compared with control subjects, specifically considering those genes involved with CSF production and CP structural integrity. Methods The Brown-Merck Gene Expression Omnibus (GEO) database (CP transcripts) was mined to examine changes in gene expression in AD compared to controls with a focus on assorted genes thought to play a role in CSF production. Specifically, genes coding for ion transporters in CP epithelium (CPE) and associated enzymes like Na–K-ATPase and carbonic anhydrase, aquaporins, mitochondrial transporters/enzymes, blood–cerebrospinal fluid barrier (BCSFB) stability proteins, and pro-inflammatory mediators were selected for investigation. Data were analyzed using t test p-value and fold-change analysis conducted by the GEO2R feature of the GEO database. Results Significant expression changes for several genes were observed in AD CP. These included disruptions to ion transporters (e.g., the solute carrier gene SLC4A5, p = 0.004) and associated enzyme expressions (e.g., carbonic anhydrase CA4, p = 0.0001), along with decreased expression of genes involved in BCSFB integrity (e.g., claudin CLDN5, p = 0.039) and mitochondrial ATP synthesis (e.g., adenosine triphosphate ATP5L, p = 0.0004). Together all changes point to disrupted solute transport at the blood–CSF interface in AD. Increased expression of pro-inflammatory (e.g., interleukin IL1RL1, p = 0.00001) and potential neurodegenerative genes (e.g., amyloid precursor APBA3, p = 0.002) also implicate disturbed CP function. Conclusions Because the altered expression of numerous transcripts in AD-CP help explain decreased CSF production in AD, these findings represent a first step towards identifying novel therapeutic targets in AD. Electronic supplementary material The online version of this article (10.1186/s12987-018-0120-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shawn Kant
- Department of Pathology (Neuropathology Division), Warren Alpert Medical School at Brown University, Providence, RI, 02903, USA
| | - Edward G Stopa
- Department of Pathology (Neuropathology Division), Warren Alpert Medical School at Brown University, Providence, RI, 02903, USA
| | - Conrad E Johanson
- Department of Neurosurgery, Warren Alpert Medical School at Brown University, Providence, RI, 02903, USA
| | - Andrew Baird
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Gerald D Silverberg
- Department of Neurosurgery, Stanford University, 710 Frenchmans Rd, Stanford, CA, 94305, USA.
| |
Collapse
|
27
|
Noda Y, Kuzuya A, Tanigawa K, Araki M, Kawai R, Ma B, Sasakura Y, Maesako M, Tashiro Y, Miyamoto M, Uemura K, Okuno Y, Kinoshita A. Fibronectin type III domain-containing protein 5 interacts with APP and decreases amyloid β production in Alzheimer's disease. Mol Brain 2018; 11:61. [PMID: 30355327 PMCID: PMC6201590 DOI: 10.1186/s13041-018-0401-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022] Open
Abstract
The deposition of Amyloid-beta peptides (Aβ) is detected at an earlier stage in Alzheimer’s disease (AD) pathology. Thus, the approach toward Aβ metabolism is considered to play a critical role in the onset and progression of AD. Mounting evidence suggests that lifestyle-related diseases are closely associated with AD, and exercise is especially linked to the prevention and the delayed progression of AD. We previously showed that exercise is more effective than diet control against Aβ pathology and cognitive deficit in AD mice fed a high-fat diet; however, the underlying molecular mechanisms remain poorly understood. On the other hand, a report suggested that exercise induced expression of fibronectin type III domain-containing protein 5 (FNDC5) in the hippocampus of mice through PGC1α pathway. Thus, in the current study, we investigated a possibility that FNDC5 interacts with amyloid precursor protein (APP) and affects Aβ metabolism. As a result, for the first time ever, we found the interaction between FNDC5 and APP, and forced expression of FNDC5 significantly decreased levels of both Aβ40 and Aβ42 secreted in the media. Taken together, our results indicate that FNDC5 significantly affects β-cleavage of APP via the interaction with APP, finally regulating Aβ levels. A deeper understanding of the mechanisms by which the interaction between APP and FNDC5 may affect Aβ production in an exercise-dependent manner would provide new preventive strategies against the development of AD.
Collapse
Affiliation(s)
- Yasuha Noda
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akira Kuzuya
- Department of Neurology, Graduated school of Medicine, Kyoto University, 54 Shogoin kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kyousuke Tanigawa
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Mitsugu Araki
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ryoko Kawai
- Department of Pharmaceuticals, Kyoto University, 46 Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Biao Ma
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe, Foundation for Biomedical Research and Innovation (FBRI), 6-3-5, Minatojima-Minamimachi Chuo-ku, Kobe, 650-0047, Japan
| | - Yoko Sasakura
- Research and Development Group for In Silico Drug Discovery, Pro-Cluster Kobe, Foundation for Biomedical Research and Innovation (FBRI), 6-3-5, Minatojima-Minamimachi Chuo-ku, Kobe, 650-0047, Japan
| | - Masato Maesako
- Neurobiology of Alzheimer's Disease Laboratory, Massachusetts General Hospital, Harvard Medical School, 149 13th Street Charlestown, Charlestown, MA, 02129, USA
| | - Yoshitaka Tashiro
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masakazu Miyamoto
- Department of Neurology, Graduated school of Medicine, Kyoto University, 54 Shogoin kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kengo Uemura
- Department of Neurology, Graduated school of Medicine, Kyoto University, 54 Shogoin kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasushi Okuno
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ayae Kinoshita
- Department of Human Health Sciences, Graduated school of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
28
|
Hladky SB, Barrand MA. Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier. Fluids Barriers CNS 2018; 15:30. [PMID: 30340614 PMCID: PMC6194691 DOI: 10.1186/s12987-018-0113-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
This review considers efflux of substances from brain parenchyma quantified as values of clearances (CL, stated in µL g-1 min-1). Total clearance of a substance is the sum of clearance values for all available routes including perivascular pathways and the blood-brain barrier. Perivascular efflux contributes to the clearance of all water-soluble substances. Substances leaving via the perivascular routes may enter cerebrospinal fluid (CSF) or lymph. These routes are also involved in entry to the parenchyma from CSF. However, evidence demonstrating net fluid flow inwards along arteries and then outwards along veins (the glymphatic hypothesis) is still lacking. CLperivascular, that via perivascular routes, has been measured by following the fate of exogenously applied labelled tracer amounts of sucrose, inulin or serum albumin, which are not metabolized or eliminated across the blood-brain barrier. With these substances values of total CL ≅ 1 have been measured. Substances that are eliminated at least partly by other routes, i.e. across the blood-brain barrier, have higher total CL values. Substances crossing the blood-brain barrier may do so by passive, non-specific means with CLblood-brain barrier values ranging from < 0.01 for inulin to > 1000 for water and CO2. CLblood-brain barrier values for many small solutes are predictable from their oil/water partition and molecular weight. Transporters specific for glucose, lactate and many polar substrates facilitate efflux across the blood-brain barrier producing CLblood-brain barrier values > 50. The principal route for movement of Na+ and Cl- ions across the blood-brain barrier is probably paracellular through tight junctions between the brain endothelial cells producing CLblood-brain barrier values ~ 1. There are large fluxes of amino acids into and out of the brain across the blood-brain barrier but only small net fluxes have been observed suggesting substantial reuse of essential amino acids and α-ketoacids within the brain. Amyloid-β efflux, which is measurably faster than efflux of inulin, is primarily across the blood-brain barrier. Amyloid-β also leaves the brain parenchyma via perivascular efflux and this may be important as the route by which amyloid-β reaches arterial walls resulting in cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
29
|
Martín-Láez R, Valle-San Román N, Rodríguez-Rodríguez E, Marco-de Lucas E, Berciano Blanco J, Vázquez-Barquero A. Current concepts on the pathophysiology of idiopathic chronic adult hydrocephalus: Are we facing another neurodegenerative disease? NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2016.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
30
|
Bors L, Tóth K, Tóth EZ, Bajza Á, Csorba A, Szigeti K, Máthé D, Perlaki G, Orsi G, Tóth GK, Erdő F. Age-dependent changes at the blood-brain barrier. A Comparative structural and functional study in young adult and middle aged rats. Brain Res Bull 2018. [PMID: 29522862 DOI: 10.1016/j.brainresbull.2018.03.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Decreased beta-amyloid clearance in Alzheimer's disease and increased blood-brain barrier permeability in aged subjects have been reported in several articles. However, morphological and functional characterization of blood-brain barrier and its membrane transporter activity have not been described in physiological aging yet. The aim of our study was to explore the structural changes in the brain microvessels and possible functional alterations of P-glycoprotein at the blood-brain barrier with aging. Our approach included MR imaging for anatomical orientation in middle aged rats, electronmicroscopy and immunohistochemistry to analyse the alterations at cellular level, dual or triple-probe microdialysis and SPECT to test P-glycoprotein functionality in young and middle aged rats. Our results indicate that the thickness of basal lamina increases, the number of tight junctions decreases and the size of astrocyte endfeet extends with advanced age. On the basis of microdialysis and SPECT results the P-gp function is reduced in old rats. With our multiparametric approach a complex regulation can be suggested which includes elements leading to increased permeability of blood-brain barrier by enhanced paracellular and transcellular transport, and factors working against it. To verify the role of P-gp pumps in brain aging further studies are warranted.
Collapse
Affiliation(s)
- Luca Bors
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest
| | - Kinga Tóth
- Hungarian Academy of Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar tudósok körútja 2. H-1117 Budapest
| | - Estilla Zsófia Tóth
- Hungarian Academy of Sciences, Institute of Cognitive Neuroscience and Psychology, Magyar tudósok körútja 2. H-1117 Budapest
| | - Ágnes Bajza
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest
| | - Attila Csorba
- University of Szeged, Faculty of Pharmacy, Deparment of Pharmacognosy, Eötvös u. 6, H-6720 Szeged
| | - Krisztián Szigeti
- Semmelweis University, Faculty of Medicine, Department of Biophysics and Radiation Biology, Tűzoltó u. 37-47, H-1094 Budapest
| | - Domokos Máthé
- Semmelweis University, Faculty of Medicine, Department of Biophysics and Radiation Biology, Tűzoltó u. 37-47, H-1094 Budapest,; CROmed Translational Research Ltd. Budapest
| | - Gábor Perlaki
- MTA-PTE Clinical Neuroscience MR Research Group, Ret u. 2, H-7623 Pecs, Hungary; Department of Neurosurgery, University of Pecs, Medical School, Ret u. 2, H-7623 Pecs, Hungary
| | - Gergely Orsi
- MTA-PTE Clinical Neuroscience MR Research Group, Ret u. 2, H-7623 Pecs, Hungary; Department of Neurosurgery, University of Pecs, Medical School, Ret u. 2, H-7623 Pecs, Hungary
| | - Gábor K Tóth
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Franciska Erdő
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, Práter u. 50a, H-1083 Budapest.
| |
Collapse
|
31
|
Liu CB, Wang R, Yi YF, Gao Z, Chen YZ. Lycopene mitigates β-amyloid induced inflammatory response and inhibits NF-κB signaling at the choroid plexus in early stages of Alzheimer's disease rats. J Nutr Biochem 2017; 53:66-71. [PMID: 29195132 DOI: 10.1016/j.jnutbio.2017.10.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/28/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
The choroid plexus is able to modulate the cognitive function, through changes in the neuroinflammatory response and in brain immune surveillance. However, whether lycopene is involved in inflammatory responses at the choroid plexus in the early stages of Alzheimer's disease, and its molecular underpinnings are elusive. In this rat study, lycopene was used to investigate its protective effects on inflammation caused by β-amyloid. We characterized the learning and memory abilities, cytokine profiles of circulating TNF-α, IL-1β and IL-6β in the serum and the expressions of Toll like receptor 4 and nuclear factor-κB p65 mRNA and protein at the choroid plexus. The results showed that functional deficits of learning and memory in lycopene treatment groups were significantly improved compared to the control group without lycopene treatment in water maze test. The levels of serum TNF-α, IL-1β and IL-6β were significantly increased, and the expressions of TLR4 and NF-κB p65 mRNA and protein at the choroid plexus were up-regulated, indicating inflammation response was initiated following administration of Aβ1-42. After intragastric pretreatment with lycopene, inflammatory cytokines were significantly reduced and lycopene also reversed the Aβ1-42 induced up-regulation of TLR4 and NF-κB p65 mRNA and protein expressions at the choroid plexus. These results provided a novel evidence that lycopene significantly improved cognitive deficits and were accompanied by the attenuation of inflammatory injury via blocking the activation of NF-κB p65 and TLR4 expressions and production of cytokines, thereby endorsing its usefulness for diminishing β-amyloid deposition in the hippocampus tissues.
Collapse
Affiliation(s)
- Chong-Bin Liu
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China; Department of Physiology, Basic Medical Science College, Wenzhou Medical University, 325035, Wenzhou, People's Republic of China.
| | - Rui Wang
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yan-Feng Yi
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Zhen Gao
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yi-Zhu Chen
- Central Blood Station of Huzhou City, 313020, Huzhou, People's Republic of China.
| |
Collapse
|
32
|
Delaney C, Campbell M. The blood brain barrier: Insights from development and ageing. Tissue Barriers 2017; 5:e1373897. [PMID: 28956691 PMCID: PMC5788423 DOI: 10.1080/21688370.2017.1373897] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/25/2017] [Accepted: 08/26/2017] [Indexed: 12/29/2022] Open
Abstract
The blood brain barrier is a necessity for cerebral homeostasis and response to environmental insult, thus loss in functionality with age creates opportunities for disease to arise in the aged brain. Understanding how the barrier is developed and maintained throughout the earlier years of adult life can identify key processes that may have beneficial applications in the restoration of the aged brain. With an unprecedented increasing global aged population, the prevention and treatment of age-associated disorders has become a rising healthcare priority demanding novel approaches for the development of therapeutic strategies. The aging cardiovascular system has long been recognised to be a major factor in age-associated diseases such as stroke, atherosclerosis and cardiac arrest. Changes in the highly specialised cerebral vasculature may similarly drive neurodegenerative and neuropsychiatric disease.
Collapse
Affiliation(s)
- Conor Delaney
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
Storck SE, Pietrzik CU. Endothelial LRP1 - A Potential Target for the Treatment of Alzheimer's Disease : Theme: Drug Discovery, Development and Delivery in Alzheimer's Disease Guest Editor: Davide Brambilla. Pharm Res 2017; 34:2637-2651. [PMID: 28948494 DOI: 10.1007/s11095-017-2267-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022]
Abstract
The accumulation of the neurotoxin beta-amyloid (Aβ) is a major hallmark in Alzheimer's disease (AD). Aβ homeostasis in the brain is governed by its production and various clearance mechanisms. Both pathways are influenced by the ubiquitously expressed low-density lipoprotein receptor-related protein 1 (LRP1). In cerebral blood vessels, LRP1 is an important mediator for the rapid removal of Aβ from brain via transport across the blood-brain barrier (BBB). Here, we summarize recent findings on LRP1 function and discuss the targeting of LRP1 as a modulator for AD pathology and drug delivery into the brain.
Collapse
Affiliation(s)
- Steffen E Storck
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany
| | - Claus U Pietrzik
- Molecular Neurodegeneration, Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Duesbergweg 6, 55099, Mainz, Germany.
| |
Collapse
|
34
|
Osgood D, Miller MC, Messier AA, Gonzalez L, Silverberg GD. Aging alters mRNA expression of amyloid transporter genes at the blood-brain barrier. Neurobiol Aging 2017; 57:178-185. [PMID: 28654861 PMCID: PMC5728118 DOI: 10.1016/j.neurobiolaging.2017.05.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022]
Abstract
Decreased clearance of potentially toxic metabolites, due to aging changes, likely plays a significant role in the accumulation of amyloid-beta (Aβ) peptides and other macromolecules in the brain of the elderly and in the patients with Alzheimer's disease (AD). Aging is the single most important risk factor for AD development. Aβ transport receptor proteins expressed at the blood-brain barrier are significantly altered with age: the efflux transporters lipoprotein receptor-related protein 1 and P-glycoprotein are reduced, whereas the influx transporter receptor for advanced glycation end products is increased. These receptors play an important role in maintaining brain biochemical homeostasis. We now report that, in a rat model of aging, gene transcription is altered in aging, as measured by Aβ receptor gene messenger RNA (mRNA) at 3, 6, 9, 12, 15, 20, 30, and 36 months. Gene mRNA expression from isolated cerebral microvessels was measured by quantitative polymerase chain reaction. Lipoprotein receptor-related protein 1 and P-glycoprotein mRNA were significantly reduced in aging, and receptor for advanced glycation end products was increased, in parallel with the changes seen in receptor protein expression. Transcriptional changes appear to play a role in aging alterations in blood-brain barrier receptor expression and Aβ accumulation.
Collapse
Affiliation(s)
- Doreen Osgood
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Miles C Miller
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Arthur A Messier
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA
| | - Liliana Gonzalez
- Department of Computer Science and Statistics, University of Rhode Island, Kingston, RI, USA
| | - Gerald D Silverberg
- Department of Neurosurgery, Warren Alpert Medical School, Brown University, Providence, RI, USA; The Aldrich Laboratories, Rhode Island Hospital, Providence, RI, USA.
| |
Collapse
|
35
|
Ferrer I. Diversity of astroglial responses across human neurodegenerative disorders and brain aging. Brain Pathol 2017; 27:645-674. [PMID: 28804999 PMCID: PMC8029391 DOI: 10.1111/bpa.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Astrogliopathy refers to alterations of astrocytes occurring in diseases of the nervous system, and it implies the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Reactive astrocytosis refers to the response of astrocytes to different insults to the nervous system, whereas astrocytopathy indicates hypertrophy, atrophy/degeneration and loss of function and pathological remodeling occurring as a primary cause of a disease or as a factor contributing to the development and progression of a particular disease. Reactive astrocytosis secondary to neuron loss and astrocytopathy due to intrinsic alterations of astrocytes occur in neurodegenerative diseases, overlap each other, and, together with astrocyte senescence, contribute to disease-specific astrogliopathy in aging and neurodegenerative diseases with abnormal protein aggregates in old age. In addition to the well-known increase in glial fibrillary acidic protein and other proteins in reactive astrocytes, astrocytopathy is evidenced by deposition of abnormal proteins such as β-amyloid, hyper-phosphorylated tau, abnormal α-synuclein, mutated huntingtin, phosphorylated TDP-43 and mutated SOD1, and PrPres , in Alzheimer's disease, tauopathies, Lewy body diseases, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease, respectively. Astrocytopathy in these diseases can also be manifested by impaired glutamate transport; abnormal metabolism and release of neurotransmitters; altered potassium, calcium and water channels resulting in abnormal ion and water homeostasis; abnormal glucose metabolism; abnormal lipid and, particularly, cholesterol metabolism; increased oxidative damage and altered oxidative stress responses; increased production of cytokines and mediators of the inflammatory response; altered expression of connexins with deterioration of cell-to-cell networks and transfer of gliotransmitters; and worsening function of the blood brain barrier, among others. Increased knowledge of these aspects will permit a better understanding of brain aging and neurodegenerative diseases in old age as complex disorders in which neurons are not the only players.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- Institute of NeuropathologyPathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos IIIMadridSpain
| |
Collapse
|
36
|
Patel P, Shah J. Role of Vitamin D in Amyloid clearance via LRP-1 upregulation in Alzheimer's disease: A potential therapeutic target? J Chem Neuroanat 2017; 85:36-42. [PMID: 28669880 DOI: 10.1016/j.jchemneu.2017.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 05/22/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022]
Abstract
Amyloid beta (Aβ) deposition is considered to be one of the primary reason to trigger Alzheimer's disease (AD). Literature clearly suggests decline in Aβ clearance to be accountable for progression of late onset AD as compared to augmented Aβ production. There may be several pathways for Aβ clearance out of which one of the major pathway is the vascular-mediated removal of Aβ from the brain across the blood-brain barrier (BBB) via efflux pumps or receptors. Among Aβ scavenger receptors, low density lipoprotein receptor related protein (LRP-1) has been most extensively studied. LRP-1, is highly expressed in neurons and located on abluminal side of the brain capillaries whose expression decreases in AD patients which give rise to increased cerebral Aβ deposition. Recent evidences reveal that post 1,25-(OH)2D3 treatment, LRP1 expression increases significantly for both in-vivo and in-vitro studies, since Vitamin D receptors (VDR) are broadly expressed in brain. Biological actions of Vitamin D are mediated via its nuclear hormone receptor vitamin D receptor (VDR) and is found to regulate many genes. Several lines of evidence suggest that VDR deficiency/inhibition can be a potential risk factor for AD and sufficient Vitamin D supplementation is beneficial to prevent AD onset/pathology or slow down the progression of disease. The present review establishes a strong correlation between Vitamin D and LRP-1 and their possible involvement in Aβ clearance and thereby emerging as new therapeutic target.
Collapse
Affiliation(s)
- Parmi Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India.
| | - Jigna Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India.
| |
Collapse
|
37
|
Aging of cerebral white matter. Ageing Res Rev 2017; 34:64-76. [PMID: 27865980 DOI: 10.1016/j.arr.2016.11.006] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/21/2016] [Accepted: 11/04/2016] [Indexed: 12/12/2022]
Abstract
White matter (WM) occupies a large volume of the human cerebrum and is mainly composed of myelinated axons and myelin-producing glial cells. The myelinated axons within WM are the structural foundation for efficient neurotransmission between cortical and subcortical areas. Similar to neuron-enriched gray matter areas, WM undergoes a series of changes during the process of aging. WM malfunction can induce serious neurobehavioral and cognitive impairments. Thus, age-related changes in WM may contribute to the functional decline observed in the elderly. In addition, aged WM becomes more susceptible to neurological disorders, such as stroke, traumatic brain injury (TBI), and neurodegeneration. In this review, we summarize the structural and functional alterations of WM in natural aging and speculate on the underlying mechanisms. We also discuss how age-related WM changes influence the progression of various brain disorders, including ischemic and hemorrhagic stroke, TBI, Alzheimer's disease, and Parkinson's disease. Although the physiology of WM is still poorly understood relative to gray matter, WM is a rational therapeutic target for a number of neurological and psychiatric conditions.
Collapse
|
38
|
Barthélémy A, Mouchard A, Bouji M, Blazy K, Puigsegur R, Villégier AS. Glial markers and emotional memory in rats following acute cerebral radiofrequency exposures. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:25343-25355. [PMID: 27696165 DOI: 10.1007/s11356-016-7758-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Abstract
The widespread mobile phone use raises concerns on the possible cerebral effects of radiofrequency electromagnetic fields (RF EMF). Reactive astrogliosis was reported in neuroanatomical structures of adaptive behaviors after a single RF EMF exposure at high specific absorption rate (SAR, 6 W/kg). Here, we aimed to assess if neuronal injury and functional impairments were related to high SAR-induced astrogliosis. In addition, the level of beta amyloid 1-40 (Aβ 1-40) peptide was explored as a possible toxicity marker. Sprague Dawley male rats were exposed for 15 min at 0, 1.5, or 6 W/kg or for 45 min at 6 W/kg. Memory, emotionality, and locomotion were tested in the fear conditioning, the elevated plus maze, and the open field. Glial fibrillary acidic protein (GFAP, total and cytosolic fractions), myelin basic protein (MBP), and Aβ1-40 were quantified in six brain areas using enzyme-linked immunosorbent assay. According to our data, total GFAP was increased in the striatum (+114 %) at 1.5 W/kg. Long-term memory was reduced, and cytosolic GFAP was increased in the hippocampus (+119 %) and in the olfactory bulb (+46 %) at 6 W/kg (15 min). No MBP or Aβ1-40 expression modification was shown. Our data corroborates previous studies indicating RF EMF-induced astrogliosis. This study suggests that RF EMF-induced astrogliosis had functional consequences on memory but did not demonstrate that it was secondary to neuronal damage.
Collapse
Affiliation(s)
- Amélie Barthélémy
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212, 5 rue Blaise Pascal, 67084, Strasbourg, France
| | - Amandine Mouchard
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Institut des Maladies Neurodégénératives CNRS UMR5293 Université de Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - Marc Bouji
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Campus des sciences et technologies, Université Saint-Joseph, Dekwaneh, Mar Roukos, Lebanon
| | - Kelly Blazy
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Unité mixte PERITOX EA 4285-UM INERIS 01 Laboratoire Périnatalité et risques toxicologiques CHU Amiens-Picardie Hôpital, Sud Avenue Laënnec, 80 480, Salouël, France
| | - Renaud Puigsegur
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France
- Sous-direction de la police technique et scientifique, 31 Avenue Franklin Roosevelt, 69130, Ecully, France
| | - Anne-Sophie Villégier
- Institut national de l'environnement industriel et des risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP no. 2, 60550, Verneuil-en-Halatte, France.
- Unité mixte PERITOX EA 4285-UM INERIS 01 Laboratoire Périnatalité et risques toxicologiques CHU Amiens-Picardie Hôpital, Sud Avenue Laënnec, 80 480, Salouël, France.
| |
Collapse
|
39
|
Duarte AC, Hrynchak MV, Gonçalves I, Quintela T, Santos CRA. Sex Hormone Decline and Amyloid β Synthesis, Transport and Clearance in the Brain. J Neuroendocrinol 2016; 28. [PMID: 27632792 DOI: 10.1111/jne.12432] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Sex hormones (SH) are essential regulators of the central nervous system. The decline in SH levels along with ageing may contribute to compromised neuroprotection and set the grounds for neurodegeneration and cognitive impairments. In Alzheimer's disease, besides other pathological features, there is an imbalance between amyloid β (Aβ) production and clearance, leading to its accumulation in the brain of older subjects. Aβ accumulation is a primary cause for brain inflammation and degeneration, as well as concomitant cognitive decline. There is mounting evidence that SH modulate Aβ production, transport and clearance. Importantly, SH regulate most of the molecules involved in the amyloidogenic pathway, their transport across brain barriers for elimination, and their degradation in the brain interstitial fluid. This review brings together data on the regulation of Aβ production, metabolism, degradation and clearance by SH.
Collapse
Affiliation(s)
- A C Duarte
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - M V Hrynchak
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - I Gonçalves
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - T Quintela
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| | - C R A Santos
- Health Sciences Research Centre - CICS-UBI, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
40
|
Do TM, Dodacki A, Alata W, Calon F, Nicolic S, Scherrmann JM, Farinotti R, Bourasset F. Age-Dependent Regulation of the Blood-Brain Barrier Influx/Efflux Equilibrium of Amyloid-β Peptide in a Mouse Model of Alzheimer's Disease (3xTg-AD). J Alzheimers Dis 2016; 49:287-300. [PMID: 26484906 DOI: 10.3233/jad-150350] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The involvement of transporters located at the blood-brain barrier (BBB) has been suggested in the control of cerebral Aβ levels, and thereby in Alzheimer's disease (AD). However, little is known about the regulation of these transporters at the BBB in animal models of AD. In this study, we investigated the BBB expression of Aβ influx (Rage) and efflux (Abcb1-Abcg2-Abcg4-Lrp-1) transporters and cholesterol transporter (Abca1) in 3-18-month-old 3xTg-AD and control mice. The age-dependent effect of BBB transporters regulation on the brain uptake clearance (Clup) of [3H]cholesterol and [3H]Aβ1 - 40 was then evaluated in these mice, using the in situ brain perfusion technique. Our data suggest that transgenes expression led to the BBB increase in Aβ influx receptor (Rage) and decrease in efflux receptor (Lrp-1). Our data also indicate that mice have mechanisms counteracting this increased net influx. Indeed, Abcg4 and Abca1 are up regulated in 3- and 3/6-month-old 3xTg-AD mice, respectively. Our data show that the balance between the BBB influx and efflux of Aβ is maintained in 3 and 6-month-old 3xTg-AD mice, suggesting that Abcg4 and Abca1 control the efflux of Aβ through the BBB by a direct (Abcg4) or indirect (Abca1) mechanism. At 18 months, the BBB Aβ efflux is significantly increased in 3xTg-AD mice compared to controls. This could result from the significant up-regulation of both Abcg2 and Abcb1 in 3xTg-AD mice compared to control mice. Thus, age-dependent regulation of several Aβ and cholesterol transporters at the BBB could ultimately limit the brain accumulation of Aβ.
Collapse
Affiliation(s)
- Tuan Minh Do
- Laboratoire de Pharmacie Clinique et pharmacocinétique, EA 4123, Université Paris-Sud 11, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Agnès Dodacki
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Wael Alata
- Faculty of Pharmacy, Laval University, Quebec (QC), Canada
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec (QC), Canada
| | - Sophie Nicolic
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Jean-Michel Scherrmann
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| | - Robert Farinotti
- Laboratoire de Pharmacie Clinique et pharmacocinétique, EA 4123, Université Paris-Sud 11, Faculté de Pharmacie, Châtenay-Malabry, France
| | - Fanchon Bourasset
- Inserm UMR-S1144, Paris, F 75006, France.,Université Paris Descartes, Paris, France.,Université Paris Diderot, Paris, France
| |
Collapse
|
41
|
Ashok A, Rai NK, Raza W, Pandey R, Bandyopadhyay S. Chronic cerebral hypoperfusion-induced impairment of Aβ clearance requires HB-EGF-dependent sequential activation of HIF1α and MMP9. Neurobiol Dis 2016; 95:179-93. [PMID: 27431094 DOI: 10.1016/j.nbd.2016.07.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 01/25/2023] Open
Abstract
Chronic cerebral hypoperfusion (CCH) manifests Alzheimer's Disease (AD) neuropathology, marked by increased amyloid beta (Aβ). Besides, hypoxia stimulates Heparin-binding EGF-like growth factor (HB-EGF) mRNA expression in the hippocampus. However, involvement of HB-EGF in CCH-induced Aβ pathology remains unidentified. Here, using Bilateral Common Carotid Artery Occlusion mouse model, we explored the mechanism of HB-EGF regulated Aβ induction in CCH. We found that HB-EGF inhibition suppressed, while exogenous-HB-EGF triggered hippocampal Aβ, proving HB-EGF-dependent Aβ increase. We also detected that HB-EGF affected the expression of primary Aβ transporters, receptor for advanced glycation end-products (RAGE) and lipoprotein receptor-related protein-1 (LRP-1), indicating impaired Aβ clearance across the blood-brain barrier (BBB). An HB-EGF-dependent loss in BBB integrity supported impaired Aβ clearance. The effect of HB-EGF on Amyloid Precursor Protein pathway was relatively insignificant, suggesting a lesser effect on Aβ generation. Delving into BBB disruption mechanism demonstrated HB-EGF-mediated stimulation of Matrix metalloprotease-9 (MMP9), which affected BBB via HB-EGF-ectodomain shedding and epidermal growth factor receptor activation. Examining the intersection of HB-EGF-regulated pathway and hypoxia revealed HB-EGF-dependent increase in transcription factor, Hypoxia-inducible factor-1alpha (HIF1α). Further, via binding to hypoxia-responsive elements in MMP9 gene, HIF1α stimulated MMP9 expression, and therefore appeared as a prominent intermediary in HB-EGF-induced BBB damage. Overall, our study reveals the essential role of HB-EGF in triggering CCH-mediated Aβ accumulation. The proposed mechanism involves an HB-EGF-dependent HIF1α increase, generating MMP9 that stimulates soluble-HB-EGF/EGFR-induced BBB disintegration. Consequently, CCH-mediated hippocampal RAGE and LRP-1 deregulation together with BBB damage impair Aβ transport and clearance where HB-EGF plays a pivotal role.
Collapse
Affiliation(s)
- Anushruti Ashok
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR campus, Lucknow, India; Developmental Toxicology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Nagendra Kumar Rai
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR campus, Lucknow, India; Developmental Toxicology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Waseem Raza
- Developmental Toxicology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Rukmani Pandey
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR campus, Lucknow, India; Developmental Toxicology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Sanghamitra Bandyopadhyay
- Academy of Scientific and Innovative Research (AcSIR), CSIR-IITR campus, Lucknow, India; Developmental Toxicology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India.
| |
Collapse
|
42
|
Martín-Láez R, Valle-San Román N, Rodríguez-Rodríguez EM, Marco-de Lucas E, Berciano Blanco JA, Vázquez-Barquero A. Current concepts on the pathophysiology of idiopathic chronic adult hydrocephalus: Are we facing another neurodegenerative disease? Neurologia 2016; 33:449-458. [PMID: 27296497 DOI: 10.1016/j.nrl.2016.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 03/14/2016] [Accepted: 03/29/2016] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Since its description five decades ago, the pathophysiology of idiopathic chronic adult hydrocephalus (iCAH) has been traditionally related to the effect that ventricular dilatation exerts on the structures surrounding the ventricular system. However, altered cerebral blood flow, especially a reduction in the CSF turnover rate, are starting to be considered the main pathophysiological elements of this disease. DEVELOPMENT Compression of the pyramidal tract, the frontostriatal and frontoreticular circuits, and the paraventricular fibres of the superior longitudinal fasciculus have all been reported in iCAH. At the level of the corpus callosum, gliosis replaces a number of commissural tracts. Cerebral blood flow is also altered, showing a periventricular watershed region limited by the subependymal arteries and the perforating branches of the major arteries of the anterior cerebral circulation. The CSF turnover rate is decreased by 75%, leading to the reduced clearance of neurotoxins and the interruption of neuroendocrine and paracrine signalling in the CSF. CONCLUSIONS iCAH presents as a complex nosological entity, in which the effects of subcortical microangiopathy and reduced CSF turnover play a key role. According to its pathophysiology, it is simpler to think of iCAH more as a neurodegenerative disease, such as Alzheimer disease or Binswanger disease than as the classical concept of hydrocephalus.
Collapse
Affiliation(s)
- R Martín-Láez
- Servicio de Neurocirugía, Hospital Universitario «Marqués de Valdecilla», Santander, Cantabria, España.
| | - N Valle-San Román
- Servicio de Radiología, Hospital Universitario «Marqués de Valdecilla», Santander, Cantabria, España
| | - E M Rodríguez-Rodríguez
- Servicio de Neurología, Hospital Universitario «Marqués de Valdecilla», Instituto de Investigación Sanitaria IDIVAL, Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Cantabria, Santander, Cantabria, España
| | - E Marco-de Lucas
- Servicio de Radiología, Hospital Universitario «Marqués de Valdecilla», Santander, Cantabria, España
| | - J A Berciano Blanco
- Servicio de Neurología, Hospital Universitario «Marqués de Valdecilla», Instituto de Investigación Sanitaria IDIVAL, Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Universidad de Cantabria, Santander, Cantabria, España
| | - A Vázquez-Barquero
- Servicio de Neurocirugía, Hospital Universitario «Marqués de Valdecilla», Santander, Cantabria, España
| |
Collapse
|
43
|
The effect of aging on brain barriers and the consequences for Alzheimer’s disease development. Mamm Genome 2016; 27:407-20. [DOI: 10.1007/s00335-016-9637-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/20/2016] [Indexed: 12/31/2022]
|
44
|
Nelson AR, Sweeney MD, Sagare AP, Zlokovic BV. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:887-900. [PMID: 26705676 PMCID: PMC4821735 DOI: 10.1016/j.bbadis.2015.12.016] [Citation(s) in RCA: 386] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/10/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Vascular insults can initiate a cascade of molecular events leading to neurodegeneration, cognitive impairment, and dementia. Here, we review the cellular and molecular mechanisms in cerebral blood vessels and the pathophysiological events leading to cerebral blood flow dysregulation and disruption of the neurovascular unit and the blood-brain barrier, which all may contribute to the onset and progression of dementia and Alzheimer's disease (AD). Particularly, we examine the link between neurovascular dysfunction and neurodegeneration including the effects of AD genetic risk factors on cerebrovascular functions and clearance of Alzheimer's amyloid-β peptide toxin, and the impact of vascular risk factors, environment, and lifestyle on cerebral blood vessels, which in turn may affect synaptic, neuronal, and cognitive functions. Finally, we examine potential experimental treatments for dementia and AD based on the neurovascular model, and discuss some critical questions to be addressed by future studies. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Amy R Nelson
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Melanie D Sweeney
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Abhay P Sagare
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
45
|
Tábuas-Pereira M, Baldeiras I, Duro D, Santiago B, Ribeiro MH, Leitão MJ, Oliveira C, Santana I. Prognosis of Early-Onset vs. Late-Onset Mild Cognitive Impairment: Comparison of Conversion Rates and Its Predictors. Geriatrics (Basel) 2016; 1:E11. [PMID: 31022805 PMCID: PMC6371125 DOI: 10.3390/geriatrics1020011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 04/03/2016] [Accepted: 04/18/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Despite having the same histopathological characteristics, early-onset and late-onset Alzheimer's disease (AD) patients show some distinct clinical and neuropsychological profiles. Early Onset Mild Cognitive Impairment (EOMCI) is a less characterized group. The aim of this study is to characterize MCI probably due to AD in terms of the clinical, genetic, Cerebrospinal fluid (CSF) biomarkers profile and conversion rate of EOMCI, compared to the late-onset form (LOMCI). METHODS 159 MCI patients were divided in two groups: 52 EOMCI (onset < 65 years) and 107 LOMCI (onset ≥ 65 years). We investigated differences in neuropsychological scores, clinical variables, ApoE genotype, CSF biomarkers (Aβ42, t-Tau and p-Tau) in both groups. Conversion was ascertained during follow-up. RESULTS EOMCI showed a longer duration of symptoms prior to the first evaluation (EOMCI = 4.57 vs. LOMCI = 3.31, p = 0.008) and scored higher on the subjective memory complaints scale (9.91 vs. 7.85, p = 0.008), but performed better in brief cognitive tests (27.81 vs. 26.51, p < 0.001 in Mini-Mental State Examination; 19.84 vs. 18.67, p = 0.005 in Montreal Cognitive Assessment) than LOMCI. ApoE genotype distribution and CSF biomarker profile were similar in both groups, as was the conversion risk. Lower Aβ42 (Hazard ratio (HR): 0.998, 95% Confidence Interval (CI) = [0.996⁻1.000], p = 0.042), higher t-Tau levels (HR: 1.003, 95%CI = [1.000⁻1.005], p = 0.039) and higher scores in the Alzheimer Disease Assessment Scale-Cognitive (HR: 1.186, 95%CI = [1.083⁻1.299], p = 0.002) increased the risk of conversion. DISCUSSION Despite differences in memory performance and memory complaints, EOMCI and LOMCI seem to represent indistinct biological groups that do not have a higher risk of conversion to AD or differ in risk factors for conversion.
Collapse
Affiliation(s)
- Miguel Tábuas-Pereira
- Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
| | - Inês Baldeiras
- Neurochemistry laboratory, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Center for Neuroscience and Cell Biology, Coimbra, Coimbra 3000-075, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Diana Duro
- Neuropsychology unit, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
| | - Beatriz Santiago
- Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Center for Neuroscience and Cell Biology, Coimbra, Coimbra 3000-075, Portugal.
- Neuropsychology unit, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
| | - Maria Helena Ribeiro
- Neurochemistry laboratory, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Maria João Leitão
- Neurochemistry laboratory, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Center for Neuroscience and Cell Biology, Coimbra, Coimbra 3000-075, Portugal.
| | - Catarina Oliveira
- Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Center for Neuroscience and Cell Biology, Coimbra, Coimbra 3000-075, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Isabel Santana
- Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Center for Neuroscience and Cell Biology, Coimbra, Coimbra 3000-075, Portugal.
- Neuropsychology unit, Neurology department, Centro Hospitalar e Universitário de Coimbra, Coimbra 3000-075, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| |
Collapse
|
46
|
Wostyn P, De Groot V, Van Dam D, Audenaert K, Killer HE, De Deyn PP. Fast circulation of cerebrospinal fluid: an alternative perspective on the protective role of high intracranial pressure in ocular hypertension. Clin Exp Optom 2015; 99:213-8. [PMID: 26691953 DOI: 10.1111/cxo.12332] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/31/2015] [Accepted: 06/23/2015] [Indexed: 12/31/2022] Open
Abstract
As ocular hypertension refers to a condition in which the intraocular pressure is consistently elevated but without development of glaucoma, study of it may provide important clues to factors that may play a protective role in glaucoma. β-amyloid, one of the key histopathological findings in Alzheimer's disease, has been reported to increase by chronic elevation of intraocular pressure in animals with experimentally induced ocular hypertension and to cause retinal ganglion cell death, pointing to similarities in molecular cell death mechanisms between glaucoma and Alzheimer's disease. On the other hand, recent studies have reported that intracranial pressure is higher in patients with ocular hypertension compared with controls, giving rise to the idea that elevated intracranial pressure may provide a protective effect for the optic nerve by decreasing the trans-lamina cribrosa pressure difference. The speculation that the higher intracranial pressure reported in ocular hypertension patients may protect against glaucoma mainly through a lower trans-lamina cribrosa pressure difference remains at least questionable. Here, we present an alternative viewpoint, according to which the protective effect of higher intracranial pressure could be due, at least in part, to a pressure-independent mechanism, namely faster cerebrospinal fluid production leading to increased cerebrospinal fluid turnover with enhanced removal of potentially neurotoxic waste products that accumulate in the optic nerve. This suggests a new hypothesis for glaucoma, which, just like Alzheimer's disease, may be considered then as an imbalance between production and clearance of neurotoxins, including β-amyloid. If confirmed, then strategies to improve cerebrospinal fluid flow are reasonable and could provide a new therapeutic approach for stopping the neurotoxic β-amyloid pathway in glaucoma.
Collapse
Affiliation(s)
- Peter Wostyn
- Department of Psychiatry, PC Sint-Amandus, Beernem, Belgium.
| | - Veva De Groot
- Department of Ophthalmology, Antwerp University Hospital, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Department of Biomedical Sciences, Antwerp, Belgium
| | - Kurt Audenaert
- Department of Psychiatry, Ghent University Hospital, Ghent, Belgium
| | | | - Peter Paul De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Department of Biomedical Sciences, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Middelheim General Hospital, Antwerp, Belgium.,Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
47
|
Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease--A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 2015; 82:593-606. [PMID: 26311408 DOI: 10.1016/j.nbd.2015.08.014] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/23/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022] Open
Abstract
Late-onset dementia is a major health concern in the ageing population. Alzheimer's disease (AD) accounts for the largest proportion (65-70%) of dementia cases in the older population. Despite considerable research effort, the pathogenesis of late-onset AD remains unclear. Substantial evidence suggests that the neurodegenerative process is initiated by chronic cerebral hypoperfusion (CCH) caused by ageing and cardiovascular conditions. CCH causes reduced oxygen, glucose and other nutrient supply to the brain, with direct damage not only to the parenchymal cells, but also to the blood-brain barrier (BBB), a key mediator of cerebral homeostasis. BBB dysfunction mediates the indirect neurotoxic effects of CCH by promoting oxidative stress, inflammation, paracellular permeability, and dysregulation of nitric oxide, a key regulator of regional blood flow. As such, BBB dysfunction mediates a vicious circle in which cerebral perfusion is reduced further and the neurodegenerative process is accelerated. Endothelial interaction with pericytes and astrocytes could also play a role in the process. Reciprocal interactions between vascular dysfunction and neurodegeneration could further contribute to the development of the disease. A comprehensive overview of the complex scenario of interacting endothelium-mediated processes is currently lacking, and could prospectively contribute to the identification of adequate therapeutic interventions. This study reviews the current literature of in vitro and ex vivo studies on endothelium-mediated mechanisms underlying vascular dysfunction in AD pathogenesis, with the aim of presenting a comprehensive overview of the complex network of causative relationships. Particular emphasis is given to vicious circles which can accelerate the process of neurovascular degeneration.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK.
| | - Annalena Venneri
- Department of Neuroscience, Medical School, University of Sheffield, Sheffield, UK; IRCCS San Camillo Foundation Hospital, Venice, Italy
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, UK
| | - Alberto Marzo
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
48
|
Gondard E, Chau HN, Mann A, Tierney TS, Hamani C, Kalia SK, Lozano AM. Rapid Modulation of Protein Expression in the Rat Hippocampus Following Deep Brain Stimulation of the Fornix. Brain Stimul 2015; 8:1058-64. [PMID: 26321354 DOI: 10.1016/j.brs.2015.07.044] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The forniceal area is currently being evaluated as a target for deep brain stimulation (DBS) to improve cognitive function in patients with Alzheimer's disease. The molecular changes at downstream targets within the stimulated circuit are unknown. OBJECTIVE To analyze the modulation of hippocampal protein expression following 1 h of fornix DBS in the rat. METHODS Animals underwent bilateral forniceal DBS for 1 h and sacrificed at different time-points after the initiation of the stimulation (1 h, 2.5 h, 5 h, 25 h). Bilateral hippocampi were isolated for western blot analyses. RESULTS Forniceal DBS led to a dramatic elevation of cFos post-stimulation, suggesting that forniceal DBS activates the hippocampus. There was also a significant increase in candidate proteins including several trophic factors, such as brain derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF) but not glial cell-derived neurotrophic factor (GDNF). There was in addition, increased expression of the synaptic markers growth associated protein 43 (GAP-43), synaptophysin and α-synuclein. No changes were observed at the studied time-points in Alzheimer's-related proteins including amyloid precursor protein (APP), tau, phosphorylated tau (ptau), or selected chaperone proteins (HSP40, HSP70 and CHIP). CONCLUSIONS Forniceal DBS triggers hippocampal activity and rapidly modulate the expression of neurotrophic factors and markers of synaptic plasticity known to play key roles in memory processing. The clinical effects of DBS of the fornix may, in part, be mediated by producing changes in the expression of these proteins.
Collapse
Affiliation(s)
- Elise Gondard
- Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada
| | - Hien N Chau
- Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada
| | - Amandeep Mann
- Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada
| | - Travis S Tierney
- Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Clement Hamani
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada; Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Suneil K Kalia
- Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| | - Andres M Lozano
- Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, 60 Leonard Street, Toronto, ON, M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada.
| |
Collapse
|
49
|
Kitaguchi N, Hasegawa M, Ito S, Kawaguchi K, Hiki Y, Nakai S, Suzuki N, Shimano Y, Ishida O, Kushimoto H, Kato M, Koide S, Kanayama K, Kato T, Ito K, Takahashi H, Mutoh T, Sugiyama S, Yuzawa Y. A prospective study on blood Aβ levels and the cognitive function of patients with hemodialysis: a potential therapeutic strategy for Alzheimer’s disease. J Neural Transm (Vienna) 2015; 122:1593-607. [DOI: 10.1007/s00702-015-1431-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 07/20/2015] [Indexed: 10/23/2022]
|
50
|
Ramanathan A, Nelson AR, Sagare AP, Zlokovic BV. Impaired vascular-mediated clearance of brain amyloid beta in Alzheimer's disease: the role, regulation and restoration of LRP1. Front Aging Neurosci 2015; 7:136. [PMID: 26236233 PMCID: PMC4502358 DOI: 10.3389/fnagi.2015.00136] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/02/2015] [Indexed: 12/12/2022] Open
Abstract
Amyloid beta (Aβ) homeostasis in the brain is governed by its production and clearance mechanisms. An imbalance in this homeostasis results in pathological accumulations of cerebral Aβ, a characteristic of Alzheimer’s disease (AD). While Aβ may be cleared by several physiological mechanisms, a major route of Aβ clearance is the vascular-mediated removal of Aβ from the brain across the blood-brain barrier (BBB). Here, we discuss the role of the predominant Aβ clearance protein—low-density lipoprotein receptor-related protein 1 (LRP1)—in the efflux of Aβ from the brain. We also outline the multiple factors that influence the function of LRP1-mediated Aβ clearance, such as its expression, shedding, structural modification and transcriptional regulation by other genes. Finally, we summarize approaches aimed at restoring LRP1-mediated Aβ clearance from the brain.
Collapse
Affiliation(s)
- Anita Ramanathan
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Amy R Nelson
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California Los Angeles, CA, USA
| |
Collapse
|