1
|
Duchesne S, Rousseau LS, Belzile-Marsolais F, Welch LA, Cournoyer B, Arseneau M, Lapierre V, Poulin SM, Potvin O, Hudon C. A Scoping Review of Alzheimers Disease Hypotheses: An Array of Uni- and Multi-Factorial Theories. J Alzheimers Dis 2024; 99:843-856. [PMID: 38788067 PMCID: PMC11191496 DOI: 10.3233/jad-230772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 05/26/2024]
Abstract
Background There is a common agreement that Alzheimers disease (AD) is inherently complex; otherwise, a general disagreement remains on its etiological underpinning, with numerous alternative hypotheses having been proposed. Objective To perform a scoping review of original manuscripts describing hypotheses and theories of AD published in the past decades. Results We reviewed 131 original manuscripts that fulfilled our inclusion criteria out of more than 13,807 references extracted from open databases. Each entry was characterized as having a single or multifactorial focus and assigned to one of 15 theoretical groupings. Impact was tracked using open citation tools. Results Three stages can be discerned in terms of hypotheses generation, with three quarter of studies proposing a hypothesis characterized as being single-focus. The most important theoretical groupings were the Amyloid group, followed by Metabolism and Mitochondrial dysfunction, then Infections and Cerebrovascular. Lately, evidence towards Genetics and especially Gut/Brain interactions came to the fore. Conclusions When viewed together, these multi-faceted reports reinforce the notion that AD affects multiple sub-cellular, cellular, anatomical, and physiological systems at the same time but at varying degree between individuals. The challenge of providing a comprehensive view of all systems and their interactions remains, alongside ways to manage this inherent complexity.
Collapse
Affiliation(s)
- Simon Duchesne
- Department of Radiology and Nuclear Medicine, Université, Laval, Quebec City, QC, Canada
| | - Louis-Simon Rousseau
- CERVO Brain Research Centre, Quebec City, QC, Canada
- School of Psychology, Université, Laval, Quebec City, QC, Canada
| | - Florence Belzile-Marsolais
- CERVO Brain Research Centre, Quebec City, QC, Canada
- School of Psychology, Université, Laval, Quebec City, QC, Canada
| | - Laurie-Ann Welch
- CERVO Brain Research Centre, Quebec City, QC, Canada
- School of Psychology, Université, Laval, Quebec City, QC, Canada
| | | | | | - Véronick Lapierre
- CERVO Brain Research Centre, Quebec City, QC, Canada
- School of Psychology, Université, Laval, Quebec City, QC, Canada
| | | | - Olivier Potvin
- Quebec Heart and Lung Research Institute, Quebec City, QC, Canada
- CERVO Brain Research Centre, Quebec City, QC, Canada
| | - Carol Hudon
- CERVO Brain Research Centre, Quebec City, QC, Canada
- School of Psychology, Université, Laval, Quebec City, QC, Canada
- VITAM Research Centre, Quebec City, QC, Canada
| |
Collapse
|
2
|
Chatanaka MK, Sohaei D, Diamandis EP, Prassas I. Beyond the amyloid hypothesis: how current research implicates autoimmunity in Alzheimer's disease pathogenesis. Crit Rev Clin Lab Sci 2023; 60:398-426. [PMID: 36941789 DOI: 10.1080/10408363.2023.2187342] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/01/2023] [Indexed: 03/23/2023]
Abstract
The amyloid hypothesis has so far been at the forefront of explaining the pathogenesis of Alzheimer's Disease (AD), a progressive neurodegenerative disorder that leads to cognitive decline and eventual death. Recent evidence, however, points to additional factors that contribute to the pathogenesis of this disease. These include the neurovascular hypothesis, the mitochondrial cascade hypothesis, the inflammatory hypothesis, the prion hypothesis, the mutational accumulation hypothesis, and the autoimmunity hypothesis. The purpose of this review was to briefly discuss the factors that are associated with autoimmunity in humans, including sex, the gut and lung microbiomes, age, genetics, and environmental factors. Subsequently, it was to examine the rise of autoimmune phenomena in AD, which can be instigated by a blood-brain barrier breakdown, pathogen infections, and dysfunction of the glymphatic system. Lastly, it was to discuss the various ways by which immune system dysregulation leads to AD, immunomodulating therapies, and future directions in the field of autoimmunity and neurodegeneration. A comprehensive account of the recent research done in the field was extracted from PubMed on 31 January 2022, with the keywords "Alzheimer's disease" and "autoantibodies" for the first search input, and "Alzheimer's disease" with "IgG" for the second. From the first search, 19 papers were selected, because they contained recent research on the autoantibodies found in the biofluids of patients with AD. From the second search, four papers were selected. The analysis of the literature has led to support the autoimmune hypothesis in AD. Autoantibodies were found in biofluids (serum/plasma, cerebrospinal fluid) of patients with AD with multiple methods, including ELISA, Mass Spectrometry, and microarray analysis. Through continuous research, the understanding of the synergistic effects of the various components that lead to AD will pave the way for better therapeutic methods and a deeper understanding of the disease.
Collapse
Affiliation(s)
- Miyo K Chatanaka
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Dorsa Sohaei
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory and Medicine Pathobiology, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
- Department of Clinical Biochemistry, University Health Network, Toronto, Canada
| | - Ioannis Prassas
- Laboratory Medicine Program, University Health Network, Toronto, Canada
| |
Collapse
|
3
|
Zhou Y, Liu X, Ma S, Zhang N, Yang D, Wang L, Ye S, Zhang Q, Ruan J, Ma J, Wang S, Jiang N, Zhao Z, Zhao S, Zheng C, Fan X, Gong Y, Abdoul Razak MY, Hu W, Pan J, Wang X, Fan J, Li J, Liu R, Shentu Y. ChK1 activation induces reactive astrogliosis through CIP2A/PP2A/STAT3 pathway in Alzheimer's disease. FASEB J 2022; 36:e22209. [PMID: 35195302 DOI: 10.1096/fj.202101625r] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Cancerous Inhibitor of PP2A (CIP2A), an endogenous PP2A inhibitor, is upregulated and causes reactive astrogliosis, synaptic degeneration, and cognitive deficits in Alzheimer's disease (AD). However, the mechanism underlying the increased CIP2A expression in AD brains remains unclear. We here demonstrated that the DNA damage-related Checkpoint kinase 1 (ChK1) is activated in AD human brains and 3xTg-AD mice. ChK1-mediated CIP2A overexpression drives inhibition of PP2A and activates STAT3, then leads to reactive astrogliosis and neurodegeneration in vitro. Infection of mouse brain with GFAP-ChK1-AAV induced AD-like cognitive deficits and exacerbated AD pathologies in vivo. In conclusion, we showed that ChK1 activation induces reactive astrogliosis, degeneration of neurons, and exacerbation of AD through the CIP2A-PP2A-STAT3 pathway, and inhibiting ChK1 may be a potential therapeutic approach for AD treatment.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | | | - Shuqing Ma
- Wenzhou Medical University, Wenzhou, China
| | - Nan Zhang
- Wenzhou Medical University, Wenzhou, China
| | | | - Ling Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Simin Ye
- Wenzhou Medical University, Wenzhou, China
| | - Qiongying Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Ruan
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Ma
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shiyi Wang
- Wenzhou Medical University, Wenzhou, China
| | - Nan Jiang
- Wenzhou Medical University, Wenzhou, China
| | | | | | - Chenfei Zheng
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mahaman Yacoubou Abdoul Razak
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital, Shenzhen University, Shenzhen, China.,Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenting Hu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingye Pan
- Key Laboratory of Critical Emergency and Artificial Intelligence of Zhejiang Province, Wenzhou, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Critical Emergency and Artificial Intelligence of Zhejiang Province, Wenzhou, China
| |
Collapse
|
4
|
Wang WZ, Li MW, Chen Y, Liu LY, Xu Y, Xia ZH, Yu Y, Wang XD, Chen W, Zhang F, Xu XY, Gao YF, Zhang JG, Qin SC, Wang H. 3×Tg-AD Mice Overexpressing Phospholipid Transfer Protein Improves Cognition Through Decreasing Amyloid-β Production and Tau Hyperphosphorylation. J Alzheimers Dis 2021; 82:1635-1649. [PMID: 34219730 DOI: 10.3233/jad-210463] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) belongs to the lipid transfer glycoprotein family. Studies have shown that it is closely related to Alzheimer's disease (AD); however, the exact effect and mechanism remain unknown. OBJECTIVE To observe the effect of PLTP overexpression on behavioral dysfunction and the related mechanisms in APP/PS1/Tau triple transgenic (3×Tg-AD) mice. METHODS AAV-PLTP-EGFP was injected into the lateral ventricle to induce PLTP overexpression. The memory of 3×Tg-AD mice and wild type (WT) mice aged 10 months were assessed using Morris water maze (MWM) and shuttle-box passive avoidance test (PAT). Western blotting and ELISA assays were used to quantify the protein contents. Hematoxylin and eosin, Nissl, and immunochemistry staining were utilized in observing the pathological changes in the brain. RESULTS 3×Tg-AD mice displayed cognitive impairment in WMW and PAT, which was ameliorated by PLTP overexpression. The histopathological hallmarks of AD, senile plaques and neurofibrillary tangles, were observed in 3×Tg-AD mice and were improved by PLTP overexpression. Besides, the increase of amyloid-β42 (Aβ42) and Aβ40 were found in the cerebral cortex and hippocampus of 3×Tg-AD mice and reversed by PLTP overexpression through inhibiting APP and PS1. PLTP overexpression also reversed tau phosphorylation at the Ser404, Thr231 and Ser199 of the hippocampus in 3×Tg-AD mice. Furthermore, PLTP overexpression induced the glycogen synthase kinase 3β (GSK3β) inactivation via upregulating GSK3β (pSer9). CONCLUSION These results suggest that PLTP overexpression has neuroprotective effects. These effects are possibly achieved through the inhibition of the Aβ production and tau phosphorylation, which is related to GSK3β inactivation.
Collapse
Affiliation(s)
- Wen-Zhi Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Ming-Wei Li
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Ying Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Li-Yuan Liu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Yong Xu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Zeng-Hui Xia
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Yang Yu
- Key Laboratory of Atherosclerosis, University of Shandong, Jinan, China.,Institute of Artherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Xiao-Dan Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Feng Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Xiao-Yan Xu
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Yong-Feng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Ji-Guo Zhang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Shu-Cun Qin
- Key Laboratory of Atherosclerosis, University of Shandong, Jinan, China.,Institute of Artherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Hao Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| |
Collapse
|
5
|
Fulop T, Tripathi S, Rodrigues S, Desroches M, Bunt T, Eiser A, Bernier F, Beauregard PB, Barron AE, Khalil A, Plotka A, Hirokawa K, Larbi A, Bocti C, Laurent B, Frost EH, Witkowski JM. Targeting Impaired Antimicrobial Immunity in the Brain for the Treatment of Alzheimer's Disease. Neuropsychiatr Dis Treat 2021; 17:1311-1339. [PMID: 33976546 PMCID: PMC8106529 DOI: 10.2147/ndt.s264910] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and aging is the most common risk factor for developing the disease. The etiology of AD is not known but AD may be considered as a clinical syndrome with multiple causal pathways contributing to it. The amyloid cascade hypothesis, claiming that excess production or reduced clearance of amyloid-beta (Aβ) and its aggregation into amyloid plaques, was accepted for a long time as the main cause of AD. However, many studies showed that Aβ is a frequent consequence of many challenges/pathologic processes occurring in the brain for decades. A key factor, sustained by experimental data, is that low-grade infection leading to production and deposition of Aβ, which has antimicrobial activity, precedes the development of clinically apparent AD. This infection is chronic, low grade, largely clinically silent for decades because of a nearly efficient antimicrobial immune response in the brain. A chronic inflammatory state is induced that results in neurodegeneration. Interventions that appear to prevent, retard or mitigate the development of AD also appear to modify the disease. In this review, we conceptualize further that the changes in the brain antimicrobial immune response during aging and especially in AD sufferers serve as a foundation that could lead to improved treatment strategies for preventing or decreasing the progression of AD in a disease-modifying treatment.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Shreyansh Tripathi
- Cluster Innovation Centre, North Campus, University of Delhi, Delhi, 110007, India.,Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
| | - Serafim Rodrigues
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain.,Mathematical Computational and Experimental Neuroscience (MCEN), BCAM - The Basque Center for Applied Mathematics, Bilbao, Spain
| | - Mathieu Desroches
- MathNeuro Team, Inria Sophia Antipolis Méditerranée, Sophia Antipolis, France.,Department of Mathematics, Université Côte d'Azur, Nice, France
| | - Ton Bunt
- Izumi Biosciences, Inc., Lexington, MA, USA
| | - Arnold Eiser
- Leonard Davis Institute, University of Pennsylvania, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Francois Bernier
- Morinaga Milk Industry Co., Ltd, Next Generation Science Institute, Kanagawa, Japan
| | - Pascale B Beauregard
- Department of Biology, Faculty of Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Annelise E Barron
- Department of Bioengineering, Stanford School of Medicine, Stanford, CA, USA
| | - Abdelouahed Khalil
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Adam Plotka
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Katsuiku Hirokawa
- Institute of Health and Life Science, Tokyo Med. Dent. University, Tokyo and Nito-Memory Nakanosogo Hospital, Department of Pathology, Tokyo, Japan
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (ASTAR), Immunos Building, Biopolis, Singapore, Singapore
| | - Christian Bocti
- Research Center on Aging, Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Benoit Laurent
- Research Center on Aging, Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
6
|
Bernstein AM, Ritch R, Wolosin JM. LOXL1 folding in exfoliation glaucoma. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 118:273-288. [PMID: 31928728 PMCID: PMC7589528 DOI: 10.1016/bs.apcsb.2019.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Exfoliation syndrome (XFS) is an age-related disease defined by the deposition of aggregated fibrous material (XFM) in the peri-cellular space. Principal morbidity occurs in the eye, where XFM accumulates on the anterior ocular tissues. GWAS have found that certain genetic variants of lysyl oxidase-like 1 (LOXL1), a matrix cross-linking enzyme that is required for elastic fiber formation confer risk for the development of XFS, but are not a single causative factor as many genetically affected individuals do not develop XFS or subsequent glaucoma (XFG). We have found that XFG cells display defects in lysosomes, microtubules, autophagy, and mitochondria resembling defects found in cells from age-related syndromes, such as the main neurodegenerative diseases. In the majority of these diseases, the determining cellular factor is a protein containing intrinsically disordered regions (IDRs) and displaying a high propensity for aggregation. We have found that in XFG patient-derived cells, LOXL1 protein is actively subjected to autophagic clearance, suggesting that LOXL1 is undergoing aggregation. In silico analysis demonstrates that LOXL1's first 369 aa constitute an IDR with the highest disorder probability peak centering around the known risk positions. Experimentally, we have found over-expression of either unmodified LOXL1 or fluorescent chimeras preserving the well-structured N-terminus cause copious intracellular aggregation and that aggregation wanes when the high IDR peaks are deleted. Overall, our work suggests that XFS/G results from the aggregation of the LOXL1 protein coupled with a reduction of cellular proteostasis capabilities in aging, resulting in a chronic build-up of LOXL1-containing protein aggregates.
Collapse
Affiliation(s)
- Audrey M. Bernstein
- SUNY Upstate Medical University, Department of Ophthalmology and Visual Sciences, Syracuse, NY, United States
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York, NY, United States
| | - J. Mario Wolosin
- Eye and Vision Research Institute, Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
7
|
Downregulation of SNX27 expression does not exacerbate amyloidogenesis in the APP/PS1 Alzheimer's disease mouse model. Neurobiol Aging 2019; 77:144-153. [DOI: 10.1016/j.neurobiolaging.2019.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/01/2019] [Accepted: 01/13/2019] [Indexed: 12/20/2022]
|
8
|
Fulop T, Witkowski JM, Larbi A, Khalil A, Herbein G, Frost EH. Does HIV infection contribute to increased beta-amyloid synthesis and plaque formation leading to neurodegeneration and Alzheimer's disease? J Neurovirol 2019; 25:634-647. [PMID: 30868421 DOI: 10.1007/s13365-019-00732-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/24/2019] [Accepted: 02/01/2019] [Indexed: 10/27/2022]
Abstract
HIV infection in the combination antiretroviral therapy (cART) era has become a chronic disease with a life expectancy almost identical to those free from this infection. Concomitantly, chronic diseases such as neurodegenerative diseases have emerged as serious clinical problems. HIV-induced cognitive changes, although clinically very diverse are collectively called HIV-associated neurocognitive disorder (HAND). HAND, which until the introduction of cART manifested clinically as a subcortical disorder, is now considered primarily cognitive disorder, which makes it similar to diseases like Alzheimer's (AD) and Parkinson's disease (PD). The pathogenesis involves either the direct effects of the virus or the effect of viral proteins such as Tat, Ggp120, and Nef. These proteins are either capable of destroying neurons directly by inducing neurotoxic mediators or by initiating neuroinflammation by microglia and astrocytes. Recently, it has become recognized that HIV infection is associated with increased production of the beta-amyloid peptide (Aβ) which is a characteristic of AD. Moreover, amyloid plaques have also been demonstrated in the brains of patients suffering from HAND. Thus, the question arises whether this production of Aβ indicates that HAND may lead to AD or it is a form of AD or this increase in Aβ production is only a bystander effect. It has also been discovered that APP in HIV and its metabolic product Aβ in AD manifest antiviral innate immune peptide characteristics. This review attempts to bring together studies linking amyloid precursor protein (APP) and Aβ production in HIV infection and their possible impact on the course of HAND and AD. These data indicate that human defense mechanisms in HAND and AD are trying to contain microorganisms by antimicrobial peptides, however by employing different means. Future studies will, no doubt, uncover the relationship between HAND and AD and, hopefully, reveal novel treatment possibilities.
Collapse
Affiliation(s)
- Tamas Fulop
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada.
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdansk, Gdansk, Poland
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, Biopolis, Singapore, Singapore.,Department of Biology, Faculty of Science, University Tunis El Manar, Tunis, Tunisia
| | - Abdelouahed Khalil
- Research Center on Aging, Geriatric Division, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001, 12th Avenue North, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Georges Herbein
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, Université of Franche-Comté (UFC), University of Bourgogne France-Comté (UBFC), F-25030, Besançon, France.,Department of Virology, CHRU Besancon, F-25030, Besancon, France
| | - Eric H Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
9
|
Shi Y, Wang Y, Wei H. Dantrolene : From Malignant Hyperthermia to Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2019; 18:668-676. [PMID: 29921212 PMCID: PMC7754833 DOI: 10.2174/1871527317666180619162649] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/07/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
Dantrolene, a ryanodine receptor antagonist, is primarily known as the only clinically acceptable and effective treatment for Malignant Hyperthermia (MH). Inhibition of Ryanodine Receptor (RyR) by dantrolene decreases the abnormal calcium release from the Sarcoplasmic Reticulum (SR) or Endoplasmic Reticulum (ER), where RyR is located. Recently, emerging researches on dissociated cells, brains slices, live animal models and patients have demonstrated that altered RyR expression and function can also play a vital role in the pathogenesis of Alzheimer's Disease (AD). Therefore, dantrolene is now widely studied as a novel treatment for AD, targeting the blockade of RyR channels or another alternative pathway, such as the inhibitory effects of NMDA glutamate receptors and the effects of ER-mitochondria connection. However, the therapeutic effects are not consistent. In this review, we focus on the relationship between the altered RyR expression and function and the pathogenesis of AD, and the potential application of dantrolene as a novel treatment for the disease.
Collapse
Affiliation(s)
- Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yong Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Fulop T, Witkowski JM, Bourgade K, Khalil A, Zerif E, Larbi A, Hirokawa K, Pawelec G, Bocti C, Lacombe G, Dupuis G, Frost EH. Can an Infection Hypothesis Explain the Beta Amyloid Hypothesis of Alzheimer's Disease? Front Aging Neurosci 2018; 10:224. [PMID: 30087609 PMCID: PMC6066504 DOI: 10.3389/fnagi.2018.00224] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia. The pathological hallmarks of the disease are extracellular senile plaques composed of beta-amyloid peptide (Aβ) and intracellular neurofibrillary tangles composed of pTau. These findings led to the "beta-amyloid hypothesis" that proposes that Aβ is the major cause of AD. Clinical trials targeting Aβ in the brain have mostly failed, whether they attempted to decrease Aβ production by BACE inhibitors or by antibodies. These failures suggest a need to find new hypotheses to explain AD pathogenesis and generate new targets for intervention to prevent and treat the disease. Many years ago, the "infection hypothesis" was proposed, but received little attention. However, the recent discovery that Aβ is an antimicrobial peptide (AMP) acting against bacteria, fungi, and viruses gives increased credence to an infection hypothesis in the etiology of AD. We and others have shown that microbial infection increases the synthesis of this AMP. Here, we propose that the production of Aβ as an AMP will be beneficial on first microbial challenge but will become progressively detrimental as the infection becomes chronic and reactivates from time to time. Furthermore, we propose that host measures to remove excess Aβ decrease over time due to microglial senescence and microbial biofilm formation. We propose that this biofilm aggregates with Aβ to form the plaques in the brain of AD patients. In this review, we will develop this connection between Infection - Aβ - AD and discuss future possible treatments based on this paradigm.
Collapse
Affiliation(s)
- Tamas Fulop
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Karine Bourgade
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Abdelouahed Khalil
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Echarki Zerif
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- Singapore Immunology Network, ASTAR, Biopolis, Singapore, Singapore
| | - Katsuiku Hirokawa
- Department of Pathology, Nitobe Memorial Nakano General Hospital, Tokyo, Japan
| | - Graham Pawelec
- Department of Internal Medicine II, Center for Medical Research, University of Tübingen, Tübingen, Germany
- Health Sciences North Research Institute, Greater Sudbury, ON, Canada
| | - Christian Bocti
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Guy Lacombe
- Division of Geriatrics, Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Programme of Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H. Frost
- Department of Microbiology and Infectious Diseases, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
11
|
Ashraf GM, Baeesa SS. Investigation of Gal-3 Expression Pattern in Serum and Cerebrospinal Fluid of Patients Suffering From Neurodegenerative Disorders. Front Neurosci 2018; 12:430. [PMID: 30008660 PMCID: PMC6033997 DOI: 10.3389/fnins.2018.00430] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022] Open
Abstract
We performed this study to investigate the possibility of a definitive pattern of Galectin-3 (Gal-3) expression in the cerebrospinal fluid (CSF) and serum of Alzheimer’s disease (AD) and Amyotrophic Lateral Sclerosis (ALS) patients. In our study, we collected the CSF and serum samples of 31 AD patients, 19 ALS patients and 50 normal healthy subjects (controls). Quantitative ELISA measured Gal-3 concentrations in CSF and serum samples. A comparative analysis was performed to analyze and understand the Gal-3 expression pattern. A number of neuropsychological assessments and statistical analyses were carried out to validate our findings. Recent researches have established the role of galectins in various neurodegenerative disorders (NDDs), but a definitive pattern of galectin expression is still elusive. A significant difference was observed in serum and CSF Gal-3 concentrations between AD patients and healthy controls. The difference in serum and CSF Gal-3 concentrations between ALS patients vs. controls was lesser as compared to AD patients vs. controls. The difference in serum and CSF Gal-3 concentrations of AD vs. ALS patients was not significant. The MMSE score and serum and CSF Gal-3 concentrations in AD and ALS patients, and controls exhibited a significant positive correlation. The findings of the present study are expected to provide an insight into the definitive pattern of Gal-3 expression in AD and ALS patients, and might thus establish Gal-3 as a strong biomarker. This in turn will open up new and promising research avenues targeting the expression of galectins to modulate the progression of NDDs, and pave the way for novel therapeutic options.
Collapse
Affiliation(s)
- Ghulam M Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saleh S Baeesa
- Division of Neurosurgery, College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Ashraf J, Ahmad J, Ali A, Ul-Haq Z. Analyzing the Behavior of Neuronal Pathways in Alzheimer's Disease Using Petri Net Modeling Approach. Front Neuroinform 2018; 12:26. [PMID: 29875647 PMCID: PMC5974338 DOI: 10.3389/fninf.2018.00026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/30/2018] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's Disease (AD) is the most common neuro-degenerative disorder in the elderly that leads to dementia. The hallmark of AD is senile lesions made by abnormal aggregation of amyloid beta in extracellular space of brain. One of the challenges in AD treatment is to better understand the mechanism of action of key proteins and their related pathways involved in neuronal cell death in order to identify adequate therapeutic targets. This study focuses on the phenomenon of aggregation of amyloid beta into plaques by considering the signal transduction pathways of Calpain-Calpastatin (CAST) regulation system and Amyloid Precursor Protein (APP) processing pathways along with Ca2+ channels. These pathways are modeled and analyzed individually as well as collectively through Stochastic Petri Nets for comprehensive analysis and thorough understating of AD. The model predicts that the deregulation of Calpain activity, disruption of Calcium homeostasis, inhibition of CAST and elevation of abnormal APP processing are key cytotoxic events resulting in an early AD onset and progression. Interestingly, the model also reveals that plaques accumulation start early (at the age of 40) in life but symptoms appear late. These results suggest that the process of neuro-degeneration can be slowed down or paused by slowing down the degradation rate of Calpain-CAST Complex. In the light of this study, the suggestive therapeutic strategy might be the prevention of the degradation of Calpain-CAST complexes and the inhibition of Calpain for the treatment of neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Javaria Ashraf
- Research Center for Modeling and Simulation, National University of Sciences and Technology, Islamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling and Simulation, National University of Sciences and Technology, Islamabad, Pakistan
| | - Amjad Ali
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
13
|
Zhou Y, Wang ZF, Li W, Hong H, Chen J, Tian Y, Liu ZY. Protective effects of microRNA-330 on amyloid β-protein production, oxidative stress, and mitochondrial dysfunction in Alzheimer's disease by targeting VAV1 via the MAPK signaling pathway. J Cell Biochem 2018; 119:5437-5448. [PMID: 29369410 DOI: 10.1002/jcb.26700] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Abstract
This study aims to explore the effect of miR-330 targeting VAV1 on amyloid β-protein (Aβ) production, oxidative stress (OS), and mitochondrial dysfunction in Alzheimer's disease (AD) mice through the MAPK signaling pathway. Putative targeted gene of miR-330 was performed by a miRNA target prediction website and dual-luciferase reporter gene assay. AD mouse model was successfully established. Fourteen C57 mice were randomized into AD and control groups. The positive protein expression rate of VAV1 was measured by immunohistochemistry. Neuron cells were assigned into control, blank, negative control (NC), miR-330 mimics, miR-330 inhibitors, siRNA-VAV1, and miR-330 inhibitors + siRNA-VAV1 groups. Expression of miR-330, VAV1, ERK1, JNK1, P38MAPK, Aβ, COX, and lipoprotein receptor-related protein-1 (LRP-1) were determined using RT-qPCR and Western blotting. Colorimetry was applied to measure the levels of OS parameters of superoxide dismutase (SOD) and malondialdehyde (MDA). Aβ production in brain tissue was detected using ELISA, while that in neuron cell was measured by radioimmunoassay. MiR-330 was down-regulated in neuron cells of AD mice and VAV1 was negatively regulated by miR-330. Compared with the control group, the positive protein expression rate of VAV1 was significantly elevated in the AD group. Overexpression of miR-330 decreased the expression of VAV1, ERK1, JNK1, P38MAPK, and Aβ, but increased the expression of COX and LRP-1. AD mice revealed elevated Aβ production and MDA with decreased SOD level. The result indicates that overexpressed miR-330 targeting VAV1 through the MAPK signaling pathway reduces Aβ production and alleviates OS and mitochondrial dysfunction in AD.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Zhou-Fan Wang
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Wei Li
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Hui Hong
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Juan Chen
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Yi Tian
- Department of Neurology, The First Hospital of Changsha, Changsha, P. R. China
| | - Zhao-Yun Liu
- Department of Cardiology, Changsha Central Hospital, Changsha, P. R. China
| |
Collapse
|
14
|
Chen ZJ, Yang YF, Zhang YT, Yang DH. Dietary Total Prenylflavonoids from the Fruits of Psoralea corylifolia L. Prevents Age-Related Cognitive Deficits and Down-Regulates Alzheimer's Markers in SAMP8 Mice. Molecules 2018; 23:E196. [PMID: 29346315 PMCID: PMC6017019 DOI: 10.3390/molecules23010196] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a serious threat for the aging society. In this study, we examined the preventive effect of the total prenylflavonoids (TPFB) prepared from the dried fruits of Psoralea corylifolia L., using an age-related AD mouse model SAMP8. We found that long-term dietary TPFB at 50 mg/kg·day significantly improved cognitive performance of the SAMP8 mice in Morris water maze tests, similar to 150 mg/kg·day of resveratrol, a popular neuro-protective compound. Furthermore, TPFB treatment showed significant improvements in various AD markers in SAMP8 brains, which were restored to near control levels of the normal mice, SAMR1. TPFB significantly reduced the level of amyloid β-peptide 42 (Aβ42), inhibited hyperphosphorylation of the microtubule-associated protein Tau, induced phosphorylation of Ser9 of the glycogen synthase kinase 3β (GSK-3β), and decreased the expression of the proinflammatory cytokines TNFα, IL-6, and IL-1β. Finally, TPFB also markedly reduced the level of serum derivatives of reactive oxygen metabolites (d-ROMs), a biomarker of oxidative stress in vivo. These results showed that dietary TPFB could effectively prevent age-related cognitive deficits and AD-like neurobiochemical changes, and may have a potential role in the prevention of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhi-Jing Chen
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Yan-Fang Yang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Ying-Tao Zhang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Dong-Hui Yang
- School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
15
|
Role of the peripheral innate immune system in the development of Alzheimer's disease. Exp Gerontol 2017; 107:59-66. [PMID: 29275160 DOI: 10.1016/j.exger.2017.12.019] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/27/2017] [Accepted: 12/20/2017] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease is one of the most devastating neurodegenerative diseases. The exact cause of the disease is still not known although many scientists believe in the beta amyloid hypothesis which states that the accumulation of the amyloid peptide beta (Aβ) in brain is the initial cause which consequently leads to pathological neuroinflammation. However, it was recently shown that Aβ may have an important role in defending the brain against infections. Thus, the balance between positive and negative impact of Aβ may determine disease progression. Microglia in the brain are innate immune cells, and brain-initiated inflammatory responses reflected in the periphery suggests that Alzheimer's disease is to some extent also a systemic inflammatory disease. Greater permeability of the blood brain barrier facilitates the transport of peripheral immune cells to the brain and vice versa so that a vicious circle originating on the periphery may contribute to the development of overt clinical AD. Persistent inflammatory challenges by pathogens in the periphery, increasing with age, may also contribute to the central propagation of the pathological changes seen clinically. Therefore, the activation status of peripheral innate immune cells may represent an early biomarker of the upcoming impact on the brain. The modulation of these cells may thus become a useful mechanism for modifying disease progression.
Collapse
|
16
|
Le Page A, Garneau H, Dupuis G, Frost EH, Larbi A, Witkowski JM, Pawelec G, Fülöp T. Differential Phenotypes of Myeloid-Derived Suppressor and T Regulatory Cells and Cytokine Levels in Amnestic Mild Cognitive Impairment Subjects Compared to Mild Alzheimer Diseased Patients. Front Immunol 2017; 8:783. [PMID: 28736551 PMCID: PMC5500623 DOI: 10.3389/fimmu.2017.00783] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/21/2017] [Indexed: 12/26/2022] Open
Abstract
Alzheimer disease (AD) is the most prevalent form of dementia although the underlying cause(s) remains unknown at this time. However, neuroinflammation is believed to play an important role and suspected contributing immune parameters can be revealed in studies comparing patients at the stage of amnestic mild cognitive impairment (aMCI) to healthy age-matched individuals. A network of immune regulatory cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) maintains immune homeostasis but there are very few data on the role of these cells in AD. Here, we investigated the presence of these cells in the blood of subjects with aMCI and mild AD (mAD) in comparison with healthy age-matched controls. We also quantitated several pro- and anti-inflammatory cytokines in sera which can influence the development and activation of these cells. We found significantly higher levels of MDSCs and Tregs in aMCI but not in mAD patients, as well as higher serum IL-1β levels. Stratifying the subjects based on CMV serostatus that is known to influence multiple immune parameters showed an absence of differences between aMCI subjects compared to mAD patients and healthy controls. We suggest that the increase in MDSCs and Tregs number in aMCI subjects may have a beneficial role in modulating inflammatory processes. However, this protective mechanism may have failed in mAD patients, allowing progression of the disease. This working hypothesis obviously requires testing in future studies.
Collapse
Affiliation(s)
- Aurélie Le Page
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Garneau
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Gilles Dupuis
- Faculty of Medicine and Health Sciences, Department of Biochemistry, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H Frost
- Faculty of Medicine and Health Sciences, Department of Infectious Diseases and Microbiology, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Anis Larbi
- ASTAR, Singapore Immunology Network, Singapore, Singapore
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Graham Pawelec
- Department of Internal Medicine II, Center for Medical Research University of Tübingen, Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Tamàs Fülöp
- Faculty of Medicine and Health Sciences, Research Center on Aging, Graduate Program in Immunology, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
17
|
Bourgade K, Dupuis G, Frost EH, Fülöp T. Anti-Viral Properties of Amyloid-β Peptides. J Alzheimers Dis 2016; 54:859-878. [DOI: 10.3233/jad-160517] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Karine Bourgade
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H. Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Tamàs Fülöp
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
18
|
Bourgade K, Le Page A, Bocti C, Witkowski JM, Dupuis G, Frost EH, Fülöp T. Protective Effect of Amyloid-β Peptides Against Herpes Simplex Virus-1 Infection in a Neuronal Cell Culture Model. J Alzheimers Dis 2016; 50:1227-41. [DOI: 10.3233/jad-150652] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Karine Bourgade
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Aurélie Le Page
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Christian Bocti
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | | | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Eric H. Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Tamás Fülöp
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
19
|
Wang X, Zhang S, Lin F, Chu W, Yue S. Elevated Galectin-3 Levels in the Serum of Patients With Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2015; 30:729-32. [PMID: 23823143 PMCID: PMC10852776 DOI: 10.1177/1533317513495107] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the central nervous system. Galectin-3 (Gal-3) is characterized by a conserved sequence within the carbohydrate recognition domain. The effect of Gal-3 in AD is presently unknown. In this study, we found significantly increased Gal-3 serum levels in patients with AD compared to control participants (P=.017). There was no significant difference between patients with mild cognitive impairment (MCI) and healthy controls (P=.143) or between patients with AD and MCI (P=.688). The degree of cognitive impairment, as measured by the Mini-Mental Status Examination score, was found to have a significant correlation with the Gal-3 serum levels in all patients and healthy controls. These data suggest that Gal-3 potentially plays a role in the neuropathogenesis of AD. The Gal-3 found in serum could be a potential candidate for a biomarker panel for AD diagnosis.
Collapse
Affiliation(s)
- Xuexin Wang
- Department of Rehabilitation Medicine, Qilu Hospital of Shandong University, Ji'nan, Shandong, People's Republic of China Department of Rehabilitation Medicine, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Shuping Zhang
- Department of Clinical Laboratory, Yantai Hospital for Infectious Diseases, Yantai, Shandong, People's Republic of China
| | - Faliang Lin
- Department of Rehabilitation Medicine, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Wenzheng Chu
- Department of Neurology, Yuhuangding Hospital, Yantai, Shandong, People's Republic of China
| | - Shouwei Yue
- Department of Rehabilitation Medicine, Qilu Hospital of Shandong University, Ji'nan, Shandong, People's Republic of China
| |
Collapse
|
20
|
Caspase-dependent degradation of MDMx/MDM4 cell cycle regulatory protein in amyloid β-induced neuronal damage. Neurosci Lett 2015; 609:182-8. [PMID: 26477779 DOI: 10.1016/j.neulet.2015.10.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/07/2015] [Accepted: 10/10/2015] [Indexed: 11/23/2022]
Abstract
MDMx/MDM4 is a negative regulator of the p53 tumor suppressor protein and is necessary for survival in dividing cells. MDMx is also expressed in postmitotic neurons, with prosurvival roles that are independent of its extensively described roles in carcinogenesis. We and others have shown a role for MDMx loss in neuronal death in vitro and in vivo in several neurodegenerative diseases. Further, we have recently shown that MDMx is targeted for proteolytic degradation by calcium-dependent proteases, calpains, in neurons in vitro, and that MDMx overexpression provided partial neuroprotection in a model of HIV-associated neurodegeneration. Here, we assessed whether amyloid β (Aβ)-induced MDMx degradation occurred in Alzheimer's Disease (AD) models. Our data shows an age-dependent reduction in MDMx levels in cholinergic neurons within the cortex of adult mice expressing the swedish mutant of the amyloid precursor protein, APP in the Tg2576 murine model of AD. In vitro, Aβ treatment of primary cortical neurons led to the caspase-dependent MDMx degradation. Our findings suggest that MDMx degradation associated with neuronal death occurs via caspase activation in neurons, and that the progressive loss of MDMx protein represents a potential mechanism of Aβ-induced neuronal death during disease progression in AD.
Collapse
|
21
|
Insulin Attenuates Beta-Amyloid-Associated Insulin/Akt/EAAT Signaling Perturbations in Human Astrocytes. Cell Mol Neurobiol 2015; 36:851-864. [PMID: 26358886 DOI: 10.1007/s10571-015-0268-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 09/03/2015] [Indexed: 01/26/2023]
Abstract
The excitatory amino acid transporters 1 and 2 (EAAT1 and EAAT2), mostly located on astrocytes, are the main mediators for glutamate clearance in humans. Malfunctions of these transporters may lead to excessive glutamate accumulation and subsequent excitotoxicity to neurons, which has been implicated in many kinds of neurodegenerative disorders including Alzheimer's disease (AD). Yet, the specific mechanism of the glutamate system dysregulation remains vague. To explore whether the insulin/protein kinase B (Akt)/EAAT signaling in human astrocytes could be disturbed by beta-amyloid protein (Aβ) and be protected by insulin, we incubated HA-1800 cells with varying concentrations of Aβ1-42 oligomers and insulin. Then the alterations of several key substrates in this signal transduction pathway were determined. Our results showed that expressions of insulin receptor, phospho-insulin receptor, phospho-protein kinase B, phospho-mammalian target of rapamycin, and EAAT1 and EAAT2 were decreased by the Aβ1-42 oligomers in a dose-dependent manner (p < 0.05) and this trend could be recovered by insulin treatment (p < 0.05). However, the expressions of total Akt and mTOR were invariant (p > 0.05), and the mRNA levels of EAAT1 and EAAT2 were also unchanged (p > 0.05). Taken together, this study indicates that Aβ1-42 oligomers could cause disturbances in insulin/Akt/EAAT signaling in astrocytes, which might be responsible for AD onset and progression. Additionally, insulin can exert protective functions to the brain by modulating protein modifications or expressions.
Collapse
|
22
|
Tang W, Tam JHK, Seah C, Chiu J, Tyrer A, Cregan SP, Meakin SO, Pasternak SH. Arf6 controls beta-amyloid production by regulating macropinocytosis of the Amyloid Precursor Protein to lysosomes. Mol Brain 2015; 8:41. [PMID: 26170135 PMCID: PMC4501290 DOI: 10.1186/s13041-015-0129-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/11/2015] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the deposition of Beta-Amyloid (Aβ) peptides in the brain. Aβ peptides are generated by cleavage of the Amyloid Precursor Protein (APP) by the β − and γ − secretase enzymes. Although this process is tightly linked to the internalization of cell surface APP, the compartments responsible are not well defined. We have found that APP can be rapidly internalized from the cell surface to lysosomes, bypassing early and late endosomes. Here we show by confocal microscopy and electron microscopy that this pathway is mediated by macropinocytosis. APP internalization is enhanced by antibody binding/crosslinking of APP suggesting that APP may function as a receptor. Furthermore, a dominant negative mutant of Arf6 blocks direct transport of APP to lysosomes, but does not affect classical endocytosis to endosomes. Arf6 expression increases through the hippocampus with the development of Alzheimer’s disease, being expressed mostly in the CA1 and CA2 regions in normal individuals but spreading through the CA3 and CA4 regions in individuals with pathologically diagnosed AD. Disruption of lysosomal transport of APP reduces both Aβ40 and Aβ42 production by more than 30 %. Our findings suggest that the lysosome is an important site for Aβ production and that altering APP trafficking represents a viable strategy to reduce Aβ production.
Collapse
Affiliation(s)
- Weihao Tang
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Joshua H K Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada.
| | - Justin Chiu
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Andrea Tyrer
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Sean P Cregan
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Susan O Meakin
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Biochemistry, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Clinical Neurological Sciences, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| |
Collapse
|
23
|
Le Page A, Bourgade K, Lamoureux J, Frost E, Pawelec G, Larbi A, Witkowski JM, Dupuis G, Fülöp T. NK Cells are Activated in Amnestic Mild Cognitive Impairment but not in Mild Alzheimer’s Disease Patients. J Alzheimers Dis 2015; 46:93-107. [DOI: 10.3233/jad-143054] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Aurélie Le Page
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Karine Bourgade
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Julie Lamoureux
- Graduate Program in Physiology-Biophysics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Sherbrooke, QC, Canada
| | - Eric Frost
- Department of Microbiology and Infectiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Graham Pawelec
- Department of Internal Medicine II, Center for Medical Research University of Tübingen, Tübingen, Germany
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A-Star), 8A Biomedical Grove, Immunos, Singapore, Singapore
| | - Jacek M. Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Tamás Fülöp
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
24
|
Staging of cognitive deficits and neuropathological and ultrastructural changes in streptozotocin-induced rat model of Alzheimer’s disease. J Neural Transm (Vienna) 2015; 122:577-92. [DOI: 10.1007/s00702-015-1394-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/09/2015] [Indexed: 12/22/2022]
|
25
|
β-Amyloid peptides display protective activity against the human Alzheimer’s disease-associated herpes simplex virus-1. Biogerontology 2014; 16:85-98. [DOI: 10.1007/s10522-014-9538-8] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/28/2014] [Indexed: 12/23/2022]
|
26
|
Chang L, Li F, Chen X, Xu S, Wang C, Chen H, Wang Q. Effects of acidic oligosaccharide sugar chain on amyloid oligomer-induced impairment of synaptic plasticity in rats. Metab Brain Dis 2014; 29:683-90. [PMID: 24723110 DOI: 10.1007/s11011-014-9521-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
Soluble amyloid-β protein (Aβ) oligomers have been recognized to be early and key intermediates in Alzheimer's disease-related synaptic dysfunction. In this study, using in vitro electrophysiology, we investigated interactions of the acidic oligosaccharide sugar chain (AOSC), a marine-derived acidic oligosaccharide, with oligomeric Aβ. We found that the inhibition of long-term potentiation (LTP) induced by Aβ oligomers can be dose dependently reversed by the application of AOSC, whereas AOSC alone did not alter normal LTP induction. Interestingly, treatment with Aβ monomers with or without AOSC did not affect LTP induction. Additionally, when fresh-made Aβ was co-incubated with AOSC before in vitro testing, there was no impairment of LTP induction. The results from Western blots demonstrated that AOSC prevent the aggregation of Aβ oligomers. These findings indicate that AOSC may reverse Aβ oligomer-mediated cytotoxicity by directly disrupting the amyloid oligomer aggregation, and this action is concentration dependent. Thus, we propose that AOSC might be a potential therapeutic drug for Alzheimer's disease due to its protection against oligomeric Aβ-induced dysfunction of synaptic plasticity.
Collapse
Affiliation(s)
- Lan Chang
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical School, Ningbo University, Ningbo, 315211, China,
| | | | | | | | | | | | | |
Collapse
|
27
|
Tam JHK, Seah C, Pasternak SH. The Amyloid Precursor Protein is rapidly transported from the Golgi apparatus to the lysosome and where it is processed into beta-amyloid. Mol Brain 2014; 7:54. [PMID: 25085554 PMCID: PMC4237969 DOI: 10.1186/s13041-014-0054-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/23/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cerebral deposition of β-amyloid peptide (Aβ). Aβ is produced by sequential cleavage of the Amyloid Precursor Protein (APP) by β- and γ-secretases. Many studies have demonstrated that the internalization of APP from the cell surface can regulate Aβ production, although the exact organelle in which Aβ is produced remains contentious. A number of recent studies suggest that intracellular trafficking also plays a role in regulating Aβ production, but these pathways are relatively under-studied. The goal of this study was to elucidate the intracellular trafficking of APP, and to examine the site of intracellular APP processing. RESULTS We have tagged APP on its C-terminal cytoplasmic tail with photoactivatable Green Fluorescent Protein (paGFP). By photoactivating APP-paGFP in the Golgi, using the Golgi marker Galactosyltranferase fused to Cyan Fluorescent Protein (GalT-CFP) as a target, we are able to follow a population of nascent APP molecules from the Golgi to downstream compartments identified with compartment markers tagged with red fluorescent protein (mRFP or mCherry); including rab5 (early endosomes) rab9 (late endosomes) and LAMP1 (lysosomes). Because γ-cleavage of APP releases the cytoplasmic tail of APP including the photoactivated GFP, resulting in loss of fluorescence, we are able to visualize the cleavage of APP in these compartments. Using APP-paGFP, we show that APP is rapidly trafficked from the Golgi apparatus to the lysosome; where it is rapidly cleared. Chloroquine and the highly selective γ-secretase inhibitor, L685, 458, cause the accumulation of APP in lysosomes implying that APP is being cleaved by secretases in the lysosome. The Swedish mutation dramatically increases the rate of lysosomal APP processing, which is also inhibited by chloroquine and L685, 458. By knocking down adaptor protein 3 (AP-3; a heterotetrameric protein complex required for trafficking many proteins to the lysosome) using siRNA, we are able to reduce this lysosomal transport. Blocking lysosomal transport of APP reduces Aβ production by more than a third. CONCLUSION These data suggests that AP-3 mediates rapid delivery of APP to lysosomes, and that the lysosome is a likely site of Aβ production.
Collapse
Affiliation(s)
- Joshua HK Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Clinical Neurological Sciences, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| |
Collapse
|
28
|
Brännström K, Öhman A, Nilsson L, Pihl M, Sandblad L, Olofsson A. The N-terminal Region of Amyloid β Controls the Aggregation Rate and Fibril Stability at Low pH Through a Gain of Function Mechanism. J Am Chem Soc 2014; 136:10956-64. [DOI: 10.1021/ja503535m] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Anders Öhman
- Department
of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 85 Umeå, Sweden
| | | | | | | | | |
Collapse
|
29
|
Xu S, Guan Q, Wang C, Wei X, Chen X, Zheng B, An P, Zhang J, Chang L, Zhou W, Mody I, Wang Q. Rosiglitazone prevents the memory deficits induced by amyloid-beta oligomers via inhibition of inflammatory responses. Neurosci Lett 2014; 578:7-11. [PMID: 24933538 DOI: 10.1016/j.neulet.2014.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 05/06/2014] [Accepted: 06/05/2014] [Indexed: 01/06/2023]
Abstract
Rosiglitazone has been known to attenuate neurodegeneration in Alzheimer's disease (AD), but the underlying mechanisms remain unclear. In this study, Morris water maze test, ELISA and electrophysiological methods were used to examine the role and underling mechanisms of rosiglitazone on Aβ42 oligomer-induced memory impairments. We found that rosiglitazone attenuated Aβ42 oligomer-induced memory impairments in rats in a dose-dependent manner. The levels of inflammatory cytokines interleukin-1 beta (IL-1β) and interferon gamma (IFNγ) were significantly increased 7 days after injection of Aβ42 oligomers into the rat hippocampus. Inhibition of microglia activation prevented Aβ42 oligomer-induced increases in IL-1β and IFNγ levels. Rosiglitazone completely prevented the increase in the levels of IL-1β and IFNγ induced by Aβ42 oligomers. Treatment of hippocampal slices with the inflammatory cytokine IL-1β or IFNγ significantly inhibited the production of long-term potentiation (LTP) in the dentate gyrus. Rosiglitazone prevented the inhibitory effects of inflammatory cytokines on LTP. Thus, inhibition of inflammatory responses may be part of the mechanisms of action of rosiglitazone on preventing memory deficits induced by Aβ42 oligmers.
Collapse
Affiliation(s)
- Shujun Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Qiao Guan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Chuang Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Xiaofei Wei
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Xiaowei Chen
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Bangxu Zheng
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Pengyuan An
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Junfang Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Lan Chang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Wenhua Zhou
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China
| | - Istvan Mody
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Qinwen Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
30
|
Sheng H, Chaparro RE, Sasaki T, Izutsu M, Pearlstein RD, Tovmasyan A, Warner DS. Metalloporphyrins as therapeutic catalytic oxidoreductants in central nervous system disorders. Antioxid Redox Signal 2014; 20:2437-64. [PMID: 23706004 DOI: 10.1089/ars.2013.5413] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Metalloporphyrins, characterized by a redox-active transitional metal (Mn or Fe) coordinated to a cyclic porphyrin core ligand, mitigate oxidative/nitrosative stress in biological systems. Side-chain substitutions tune redox properties of metalloporphyrins to act as potent superoxide dismutase mimics, peroxynitrite decomposition catalysts, and redox regulators of transcription factor function. With oxidative/nitrosative stress central to pathogenesis of CNS injury, metalloporphyrins offer unique pharmacologic activity to improve the course of disease. RECENT ADVANCES Metalloporphyrins are efficacious in models of amyotrophic lateral sclerosis, Alzheimer's disease, epilepsy, neuropathic pain, opioid tolerance, Parkinson's disease, spinal cord injury, and stroke and have proved to be useful tools in defining roles of superoxide, nitric oxide, and peroxynitrite in disease progression. The most substantive recent advance has been the synthesis of lipophilic metalloporphyrins offering improved blood-brain barrier penetration to allow intravenous, subcutaneous, or oral treatment. CRITICAL ISSUES Insufficient preclinical data have accumulated to enable clinical development of metalloporphyrins for any single indication. An improved definition of mechanisms of action will facilitate preclinical modeling to define and validate optimal dosing strategies to enable appropriate clinical trial design. Due to previous failures of "antioxidants" in clinical trials, with most having markedly less biologic activity and bioavailability than current-generation metalloporphyrins, a stigma against antioxidants has discouraged the development of metalloporphyrins as CNS therapeutics, despite the consistent definition of efficacy in a wide array of CNS disorders. FUTURE DIRECTIONS Further definition of the metalloporphyrin mechanism of action, side-by-side comparison with "failed" antioxidants, and intense effort to optimize therapeutic dosing strategies are required to inform and encourage clinical trial design.
Collapse
Affiliation(s)
- Huaxin Sheng
- 1 Department of Anesthesiology, Duke University Medical Center (DUMC) , Durham, North Carolina
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang K, Zhu L, Zhu X, Zhang K, Huang B, Zhang J, Zhang Y, Zhu L, Zhou B, Zhou F. Protective effect of paeoniflorin on Aβ25-35-induced SH-SY5Y cell injury by preventing mitochondrial dysfunction. Cell Mol Neurobiol 2014; 34:227-34. [PMID: 24263411 DOI: 10.1007/s10571-013-0006-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 11/09/2013] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a major neurodegenerative brain disorder affecting about 14 million people worldwide. Aβ-induced cell injury is a crucial cause of neuronal loss in AD, thus the suppression of which might be useful for the treatment of this disease. In this study, we aimed to evaluate the effect of paeoniflorin (PF), a monoterpene glycoside isolated from aqueous extract of Radix Paeoniae Alba, on Aβ25-35-induced cytotoxicity in SH-SY5Y cells. The results showed PF could attenuate or restore the viability loss, apoptotic increase, and ROS production induced by Aβ25-35 in SH-SY5Y cells. In addition, PF strikingly inhibited Aβ25-35-induced mitochondrial dysfunction, which includes decreased mitochondrial membrane potential, increased Bax/Bcl-2 ratio, cytochrome c release and activity of caspase-3 and caspase-9. Therefore, our study provided the first experimental evidence that PF could modulate ROS production and apoptotic mitochondrial pathway in model of neuron injury in vitro and which might provide new insights into its application toward Alzheimer's disease therapy.
Collapse
Affiliation(s)
- Ke Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu Province, China,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
NF-κB mediated regulation of adult hippocampal neurogenesis: relevance to mood disorders and antidepressant activity. BIOMED RESEARCH INTERNATIONAL 2014; 2014:612798. [PMID: 24678511 PMCID: PMC3942292 DOI: 10.1155/2014/612798] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/28/2013] [Indexed: 12/18/2022]
Abstract
Adult hippocampal neurogenesis is a peculiar form of process of neuroplasticity that in recent years has gained great attention for its potential implication in cognition and in emotional behavior in physiological conditions. Moreover, a vast array of experimental studies suggested that adult hippocampal neurogenesis may be altered in various neuropsychiatric disorders, including major depression, where its disregulation may contribute to cognitive impairment and/or emotional aspects associated with those diseases. An intriguing area of interest is the potential influence of drugs on adult neurogenesis. In particular, several psychoactive drugs, including antidepressants, were shown to positively modulate adult hippocampal neurogenesis. Among molecules which could regulate adult hippocampal neurogenesis the NF-κB family of transcription factors has been receiving particular attention from our and other laboratories. Herein we review recent data supporting the involvement of NF-κB signaling pathways in the regulation of adult neurogenesis and in the effects of drugs that are endowed with proneurogenic and antidepressant activity. The potential implications of these findings on our current understanding of the process of adult neurogenesis in physiological and pathological conditions and on the search for novel antidepressants are also discussed.
Collapse
|
33
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
34
|
Prakash A, Kumar A. Role of nuclear receptor on regulation of BDNF and neuroinflammation in hippocampus of β-amyloid animal model of Alzheimer's disease. Neurotox Res 2013; 25:335-47. [PMID: 24277156 DOI: 10.1007/s12640-013-9437-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/27/2013] [Accepted: 10/29/2013] [Indexed: 12/16/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists have been reported to provide neuroprotective effects against neurodegenerative diseases. The current study was carried out to investigate the effects of chronic administration of pioglitazone, a PPAR-γ agonist, on cognitive impairment in an animal model of Alzheimer's disease induced by β-amyloid. Wistar rats received intracerebroventricular (ICV) β-amyloid (βA) application (3 nmol/3 μL), and behavioral alterations (locomotor activity and memory performance) were assessed. Animals were sacrificed immediately following the last behavioral session, and their brains were removed and dissected. Mitochondrial enzymes, oxidative parameters, inflammatory mediators (TNF-α, IL-6), caspase activity, and BDNF levels were measured in the hippocampus. ICV βA-treated rats showed a memory deficit and significantly decreased BDNF level, simultaneously, increase in mitochondrial oxidative damage and inflammatory mediators in the hippocampus. Memory impairment and oxidative damage were reversed by administration of pioglitazone (15 and 30 mg/kg). Pioglitazone also significantly restored the BDNF level and attenuated the actions of inflammatory markers in ICV βA-treated rats. However, pretreatment with PPAR-γ antagonist BADGE (15 mg/kg) with higher dose of pioglitazone significantly reversed its protective action in memory impairment in βA-treated rats, which indicates the involvement of PPAR-γ receptors mediating neuroprotective action. These results demonstrate that pioglitazone offers protection against β-amyloid-induced memory dysfunction possibly due to its antioxidant, anti-inflammatory, anti-apoptotic action and neurogenesis-like effect therefore, could have a therapeutic potential in Alzheimer's disease.
Collapse
Affiliation(s)
- Atish Prakash
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India,
| | | |
Collapse
|
35
|
Weon JB, Yun BR, Lee J, Eom MR, Ko HJ, Kim JS, Lee HY, Park DS, Chung HC, Chung JY, Ma CJ. Effect of Codonopsis lanceolata with Steamed and Fermented Process on Scopolamine-Induced Memory Impairment in Mice. Biomol Ther (Seoul) 2013; 21:405-10. [PMID: 24244829 PMCID: PMC3825205 DOI: 10.4062/biomolther.2013.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/05/2013] [Accepted: 09/02/2013] [Indexed: 11/06/2022] Open
Abstract
Codonopsis lanceolata (Campanulaceae) traditionally have been used as a tonic and to treat patients with lung abscesses. Recently, it was proposed that the extract and some compounds isolated from C. lanceolata reversed scopolamine-induced memory and learning deficits. The purpose of this study was to evaluate the improvement of cognitive enhancing effect of C. lanceolata by steam and fermentation process in scopolamine-induced memory impairment mice models by passive avoidance test and Morris water maze test. The extract of C. lanceolata or the extract of steamed and fermented C. lanceolata (SFCE) was orally administered to male mice at the doses of 100 and 300 mg/kg body weight. As a result, mice treated with steamed and fermented C. lanceolata extract (SFCE) (300 mg/kg body weight, p.o.) showed shorter escape latencies than those with C. lanceolata extract or the scopolamine-administered group in Morris water maze test. Also, it exerted longer step-through latency time than scopolamine treated group in passive avoidance test. Furthermore, neuroprotective effect of SFCE on glutamate-induced cytotoxicity was assessed in HT22 cells. Only SFCE-treated cells showed significant protection at 500 μg/ml. Interestingly, steamed C. lanceolata with fermentation contained more phenolic acid including gallic acid and vanillic acid than original C. lanceolata. Collectively, these results suggest that steam and fermentation process of C. lanceolata increased cognitive enhancing activity related to the memory processes and neuroprotective effect than original C. lanceolata.
Collapse
Affiliation(s)
- Jin Bae Weon
- Department of Medical Biomaterials Engineering, College of Biomedical Science
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kang M, Kim SY, An SSA, Ju YR. Characterizing affinity epitopes between prion protein and β-amyloid using an epitope mapping immunoassay. Exp Mol Med 2013; 45:e34. [PMID: 23907583 PMCID: PMC3789258 DOI: 10.1038/emm.2013.63] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/12/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022] Open
Abstract
Cellular prion protein, a membrane protein, is expressed in all mammals. Prion protein is also found in human blood as an anchorless protein, and this protein form is one of the many potential sources of misfolded prion protein replication during transmission. Many studies have suggested that β-amyloid1-42 oligomer causes neurotoxicity associated with Alzheimer's disease, which is mediated by the prion protein that acts as a receptor and regulates the hippocampal potentiation. The prevention of the binding of these proteins has been proposed as a possible preventative treatment for Alzheimer's disease; therefore, a greater understanding of the binding hot-spots between the two molecules is necessary. In this study, the epitope mapping immunoassay was employed to characterize binding epitopes within the prion protein and complementary epitopes in β-amyloid. Residues 23-39 and 93-119 in the prion protein were involved in binding to β-amyloid1-40 and 1-42, and monomers of this protein interacted with prion protein residues 93-113 and 123-166. Furthermore, β-amyloid antibodies against the C-terminus detected bound β-amyloid1-42 at residues 23-40, 104-122 and 159-175. β-Amyloid epitopes necessary for the interaction with prion protein were not determined. In conclusion, charged clusters and hydrophobic regions of the prion protein were involved in binding to β-amyloid1-40 and 1-42. The 3D structure appears to be necessary for β-amyloid to interact with prion protein. In the future, these binding sites may be utilized for 3D structure modeling, as well as for the pharmaceutical intervention of Alzheimer's disease.
Collapse
Affiliation(s)
- Mino Kang
- Department of Bionanotechnology, Gachon University, Gyeonggi, Korea
| | - Su Yeon Kim
- Division of Zoonoses, Center for Immunology and Pathology, National Institute of Health, Korea Centers for Disease Control and Prevention, Osong, Republic of Korea
| | - Seong Soo A An
- Department of Bionanotechnology, Gachon University, Gyeonggi, Korea
| | - Young Ran Ju
- Division of Zoonoses, Center for Immunology and Pathology, National Institute of Health, Korea Centers for Disease Control and Prevention, Osong, Republic of Korea
| |
Collapse
|
37
|
The Ameliorating Effect of Steamed and Fermented Codonopsis lanceolata on Scopolamine-Induced Memory Impairment in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:464576. [PMID: 23935665 PMCID: PMC3727085 DOI: 10.1155/2013/464576] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/11/2013] [Accepted: 06/23/2013] [Indexed: 11/18/2022]
Abstract
Codonopsis lanceolata (Campanulaceae) have been traditionally used to treat lung inflammatory diseases, such as asthma, tonsillitis, and pharyngitis. The present study was performed to evaluate the cognitive-enhancing effects of steamed and fermented C. lanceolata in scopolamine-induced memory impairments in mice. Cognitive abilities were determined by the Morris water maze and passive avoidance tests. Mice orally received fermented C. lanceolata extract at doses of 100, 300, or 500 mg/kg body weight. Fermented C. lanceolata extract (500 mg/kg body weight, p.o.) significantly shortened the escape latency times that were increased by scopolamine on the 4th day of trial sessions in the Morris water maze task. In addition, it exerted longer step-through latency times than those of the scopolamine-treated group in the passive avoidance test. Furthermore, the neuroprotective effects of fermented C. lanceolata extract on glutamate-induced neurocytotoxicity were investigated in HT22 cells. Fermented C. lanceolata extract showed a relative protection ratio of 59.62% at 500 μg/mL. In conclusion, fermented C. lanceolata extract ameliorated scopolamine-induced memory impairments, exerted neuroprotective effects, and improved activity compared to that found with original C. lanceolata. Further study will be required to investigate the mechanisms underlying this cognitive-enhancing activity.
Collapse
|
38
|
Yang HJ, Hwang JT, Kwon DY, Kim MJ, Kang S, Moon NR, Park S. Yuzu extract prevents cognitive decline and impaired glucose homeostasis in β-amyloid-infused rats. J Nutr 2013; 143:1093-9. [PMID: 23719224 DOI: 10.3945/jn.112.173401] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Our preliminary study revealed that dementia induced by β-amyloid accumulation impairs peripheral glucose homeostasis (unpublished). We therefore evaluated whether long-term oral consumption of yuzu (Citrus junos Tanaka) extract improves cognitive dysfunction and glucose homeostasis in β-amyloid-induced rats. Male rats received hippocampal CA1 infusions of β-amyloid (25-35) [plaque forming β-amyloid; Alzheimer disease (AD)] or β-amyloid (35-25) [non-plaque forming β-amyloid; C (non-Alzheimer disease control)] at a rate of 3.6 nmol/d for 14 d. AD rats were divided into 2 dietary groups that received either 3% lyophilized 70% ethanol extracts of yuzu (AD-Y) or 3% dextrin (AD-C) in high-fat diets (43% energy as fat). The AD-C group exhibited greater hippocampal β-amyloid deposition, which was not detected in the C group, and attenuated hippocampal insulin signaling. Yuzu treatment prevented β-amyloid accumulation, increased tau phosphorylation, and attenuated hippocampal insulin signaling observed in AD-C rats. Consistent with β-amyloid accumulation, the AD-C rats experienced cognitive dysfunction, which was prevented by yuzu. AD-C rats gained less weight than did C rats due to decreased feed consumption, and yuzu treatment prevented the decrease in feed consumption. Serum glucose concentrations were higher in AD-C than in C rats at 40-120 min after glucose loading during an oral-glucose-tolerance test, but not at 0-40 min. Serum insulin concentrations were highly elevated in AD-C rats but not enough to lower serum glucose to normal concentrations, indicating that rats in the AD-C group had insulin resistance and a borderline diabetic state. Although AD-C rats were profoundly insulin resistant, AD-Y rats exhibited normal first and second phases of glucose tolerance and insulin sensitivity and secretion. In conclusion, yuzu treatment prevented the cognitive dysfunction and impaired energy and glucose homeostasis induced by β-amyloid infusion.
Collapse
Affiliation(s)
- Hye Jeong Yang
- Food Certification Center, Korean Food Research Institutes, Sungnam, South Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Yan FL, Han GL, Wu GJ. Cytotoxic role of advanced glycation end-products in PC12 cells treated with β‑amyloid peptide. Mol Med Rep 2013; 8:367-72. [PMID: 23799541 DOI: 10.3892/mmr.2013.1545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/17/2013] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia afflicting the elderly. Recent studies have increasingly suggested that a high concentration of advanced glycation end products (AGEs) may be important in AD pathogenesis. However, the mechanisms and pathways involved remain unknown. The aim of this study was to explore whether the mechanism of the effect of AGEs on Aβ‑PC12 cells [PC12 cells treated with β‑amyloid (Aβ) peptide] was associated with oxidative stress; and to study whether inhibiting the activity of the receptor for AGE (RAGE) attenuated the toxic effect of AGEs and Aβ on PC12 cells. Several PC12 cells were pretreated with Aβ, and were then treated with different concentrations of AGEs. Other PC12 cells were treated with trypsin, a pancreatic protein enzyme and an inhibitor of RAGE, and were then treated with Aβ and AGEs. Apoptosis was measured by flow cytometry (FCM) and cell viability was measured by MTT assay. RAGE and nuclear factor‑κB (NF‑κB) were measured by reverse transcription-polymerase chain reaction (RT‑PCR) assay. With an increase in AGE concentration, the viability of Aβ‑PC12 cells treated with AGEs decreased. However, the Aβ‑PC12 cell viability was greater in the trypsin group than in the non‑trypsin group. Cell apoptosis rates and mRNA expression of RAGE and NF‑κB in Aβ‑PC12 cells treated with AGEs were significantly higher than in the Aβ‑PC12 cells. AGEs and Aβ were neurotoxic, and RAGE triggered the neural cytotoxic role of AGEs in Aβ‑PC12 cells. The molecular mechanisms may be connected with the expression of NF‑κB and apoptosis mediated by RAGE. Inhibiting the activity of RAGE may mitigate the toxic effect of AGEs and Aβ on neural cells.
Collapse
Affiliation(s)
- Fu-Ling Yan
- Department of Neurology, Zhongda Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China.
| | | | | |
Collapse
|
40
|
Stargardt A, Gillis J, Kamphuis W, Wiemhoefer A, Kooijman L, Raspe M, Benckhuijsen W, Drijfhout JW, M. Hol E, Reits E. Reduced amyloid-β degradation in early Alzheimer's disease but not in the APPswePS1dE9 and 3xTg-AD mouse models. Aging Cell 2013; 12:499-507. [PMID: 23534431 DOI: 10.1111/acel.12074] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2013] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is hallmarked by amyloid-β (Aβ) peptides accumulation and aggregation in extracellular plaques, preceded by intracellular accumulation. We examined whether intracellular Aβ can be cleared by cytosolic peptidases and whether this capacity is affected during progression of sporadic AD (sAD) in humans and in the commonly used APPswePS1dE9 and 3xTg-AD mouse models. A quenched Aβ peptide that becomes fluorescent upon degradation was used to screen for Aβ-degrading cytoplasmic peptidases cleaving the aggregation-prone KLVFF region of the peptide. In addition, this quenched peptide was used to analyze Aβ-degrading capacity in the hippocampus of sAD patients with different Braak stages as well as APPswePS1dE9 and 3xTg-AD mice. Insulin-degrading enzyme (IDE) was found to be the main peptidase that degrades cytoplasmic, monomeric Aβ. Oligomerization of Aβ prevents its clearance by IDE. Intriguingly, the Aβ-degrading capacity decreases already during the earliest Braak stages of sAD, and this decline correlates with IDE protein levels, but not with mRNA levels. This suggests that decreased IDE levels could contribute to early sAD. In contrast to the human data, the commonly used APPswePS1dE9 and 3xTg-AD mouse models do not show altered Aβ degradation and IDE levels with AD progression, raising doubts whether mouse models that overproduce Aβ peptides are representative for human sAD.
Collapse
Affiliation(s)
- Anita Stargardt
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Judith Gillis
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Willem Kamphuis
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
| | - Anne Wiemhoefer
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Lieneke Kooijman
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
| | - Marcel Raspe
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| | - Willemien Benckhuijsen
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Albinusdreef 2, 2333 ZA Leiden The Netherlands
| | - Jan W. Drijfhout
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Albinusdreef 2, 2333 ZA Leiden The Netherlands
| | - Elly M. Hol
- Department of Astrocyte Biology and Neurodegeneration Netherlands Institute for Neuroscience – an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW) Meibergdreef 47, 1105 BA Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences Center for Neuroscience University of Amsterdam Sciencepark 904 1098 XH AmsterdamThe Netherlands
| | - Eric Reits
- Department of Cell Biology and Histology Academic Medical Center Meibergdreef 151105 AZ Amsterdam The Netherlands
| |
Collapse
|
41
|
Kumar S, Wirths O, Theil S, Gerth J, Bayer TA, Walter J. Early intraneuronal accumulation and increased aggregation of phosphorylated Abeta in a mouse model of Alzheimer's disease. Acta Neuropathol 2013; 125:699-709. [PMID: 23525537 DOI: 10.1007/s00401-013-1107-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/06/2013] [Accepted: 03/09/2013] [Indexed: 12/13/2022]
Abstract
The progressive accumulation of extracellular amyloid plaques in the brain is a common hallmark of Alzheimer's disease (AD). We recently identified a novel species of Aβ phosphorylated at serine residue 8 with increased propensity to form toxic aggregates as compared to non-phosphorylated species. The age-dependent analysis of Aβ depositions using novel monoclonal phosphorylation-state specific antibodies revealed that phosphorylated Aβ variants accumulate first inside of neurons in a mouse model of AD already at 2 month of age. At higher ages, phosphorylated Aβ is also abundantly detected in extracellular plaques. Besides a large overlap in the spatiotemporal deposition of phosphorylated and non-phosphorylated Aβ species, fractionized extraction of Aβ from brains revealed an increased accumulation of phosphorylated Aβ in oligomeric assemblies as compared to non-phosphorylated Aβ in vivo. Thus, phosphorylated Aβ could represent an important species in the formation and stabilization of neurotoxic aggregates, and might be targeted for AD therapy and diagnosis.
Collapse
Affiliation(s)
- Sathish Kumar
- Department of Neurology, University of Bonn, 53127, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Koh DC, Edelman GM, Mauro VP. Physical evidence supporting a ribosomal shunting mechanism of translation initiation for BACE1 mRNA. ACTA ACUST UNITED AC 2013; 1:e24400. [PMID: 26824018 PMCID: PMC4718059 DOI: 10.4161/trla.24400] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 03/06/2013] [Accepted: 03/21/2013] [Indexed: 12/22/2022]
Abstract
In Alzheimer disease, elevated levels of the BACE1 enzyme are correlated with increased production of amyloid peptides and disease pathology. The increase in BACE1 levels is post-transcriptional and may involve altered translation efficiency. Earlier studies have indicated that translation of BACE1 mRNA is cap-dependent. As ribosomal subunits move from the cap-structure to the initiation codon, they fail to recognize several AUG codons in the 5′ leader. In this study, we looked for physical evidence of the mechanism underlying ribosomal scanning or shunting along the BACE1 5′ leader by investigating structural stability in the 5′ leaders of endogenous mRNAs in vivo. To perform this analysis, we probed RNAs using lead(II) acetate, a cell-permeable chemical that induces cleavage of unpaired nucleotides having conformational flexibility. The data revealed that the ≈440-nt 5′ leader was generally resistant to cleavage except for a region upstream of the initiation codon. Cleavage continued into the coding region, consistent with destabilization of secondary structures by translating ribosomes. Evidence that a large segment of the BACE1 5′ leader was not cleaved indicates that this region is structurally stable and suggests that it is not scanned. The data support a mechanism of translation initiation in which ribosomal subunits bypass (shunt) part of the BACE1 5′ leader to reach the initiation codon. We suggest that a nucleotide bias in the 5′ leader may predispose the initiation codon to be more accessible than other AUG codons in the 5′ leader, leading to an increase in its relative utilization.
Collapse
Affiliation(s)
- Dora C Koh
- Department of Cell and Molecular Biology; The Scripps Research Institute; La Jolla, CA USA
| | - Gerald M Edelman
- Department of Cell and Molecular Biology; The Scripps Research Institute; La Jolla, CA USA
| | - Vincent P Mauro
- Department of Cell and Molecular Biology; The Scripps Research Institute; La Jolla, CA USA
| |
Collapse
|
43
|
Ma WW, Hou CC, Zhou X, Yu HL, Xi YD, Ding J, Zhao X, Xiao R. Genistein alleviates the mitochondria-targeted DNA damage induced by β-amyloid peptides 25–35 in C6 glioma cells. Neurochem Res 2013; 38:1315-23. [DOI: 10.1007/s11064-013-1019-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/07/2013] [Accepted: 03/12/2013] [Indexed: 12/21/2022]
|
44
|
Brasnjevic I, Lardenoije R, Schmitz C, Van Der Kolk N, Dickstein DL, Takahashi H, Hof PR, Steinbusch HW, Rutten BP. REGION-SPECIFIC NEURON AND SYNAPSE LOSS IN THE HIPPOCAMPUS OF APP SL/PS1 KNOCK-IN MICE. Transl Neurosci 2013; 4:8-19. [PMID: 24829793 PMCID: PMC4018205 DOI: 10.2478/s13380-013-0111-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Transgenic mouse models with knock-in (KI) expression of human mutant amyloid precursor protein (APP) and/or human presenilin 1 (PS1) may be helpful to elucidate the cellular consequences of APP and PS1 misprocessing in the aging brain. Age-related alterations in total numbers of neurons and in numbers of synaptophysin-immunoreactive presynaptic boutons (SIPB), as well as the amyloid plaque load were analyzed in the hippocampal dentate gyrus (DG), CA3, and CA1-2 of 2- and 10-month-old APPSL/PS1 homozygous KI, APPSL (expressing human mutant APP751 carrying the Swedish [K670N/M671L] and London [V717I] mutations under Thy-1 promoter), and PS1 homozygous KI mice (expressing human PS1 mutations [M233T and L235P]). APPSL/PS1 homozygous KI mice, but neither APPSL mice nor PS1 homozygous KI mice, showed substantial age-related loss of neurons (-47.2%) and SIPB (-22.6%), specifically in CA1-2. PS1 homozygous KI mice showed an age-related increase in hippocampal granule cell numbers (+37.9%). Loss of neurons and SIPB greatly exceeded the amount of local extracellular Aβ aggregation and astrocytes, whereas region-specific accumulation of intraneuronal Aβ preceded neuron and synapse loss. An age-related increase in the ratio of SIPB to neuron numbers in CA1-2 of APPSL/PS1 homozygous KI mice was suggestive of compensatory synaptic plasticity. These findings indicate a region-selectivity in intra- and extraneuronal Aβ accumulation in connection with neuron and synapse loss in the hippocampus of APPSL/PS1 homozygous KI mice.
Collapse
Affiliation(s)
- Ivona Brasnjevic
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience (EURON), 6200 MD Maastricht, The Netherlands
| | - Roy Lardenoije
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Christoph Schmitz
- Department of Anatomy II, Institute of Anatomy, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Nicolien Van Der Kolk
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience (EURON), 6200 MD Maastricht, The Netherlands
| | - Dara L. Dickstein
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hisaaki Takahashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, 791-0295 Ehime, Japan
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Harry W.M. Steinbusch
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience (EURON), 6200 MD Maastricht, The Netherlands
| | - Bart P.F. Rutten
- Department of Psychiatry and Neuropsychology, Maastricht University, 6200 MD Maastricht, The Netherlands
- European Graduate School of Neuroscience (EURON), 6200 MD Maastricht, The Netherlands
| |
Collapse
|
45
|
Carrillo MC, Brashear HR, Logovinsky V, Ryan JM, Feldman HH, Siemers ER, Abushakra S, Hartley DM, Petersen RC, Khachaturian AS, Sperling RA. Can we prevent Alzheimer's disease? Secondary "prevention" trials in Alzheimer's disease. Alzheimers Dement 2013; 9:123-131.e1. [PMID: 23411394 DOI: 10.1016/j.jalz.2012.12.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Current research including the basic biology of Alzheimer's disease (AD) provides a foundation to explore whether our current state of knowledge is sufficient to initiate prevention studies and allow us to believe prevention of AD is possible. Current research and recently revised criteria for the diagnosis of AD by the National Institutes on Aging and the Alzheimer's Association suggest a continuum of disease from preclinical asymptomatic to symptomatic Alzheimer's dementia. In light of these revised criteria, the possibility of secondary prevention and even primary prevention is under discussion. The Alzheimer's Association Research Roundtable convened a meeting to discuss the rationale and feasibility of conducting secondary prevention trials in AD.
Collapse
|
46
|
Anti-amyloid-β-mediated positron emission tomography imaging in Alzheimer's disease mouse brains. PLoS One 2012; 7:e51958. [PMID: 23284831 PMCID: PMC3528731 DOI: 10.1371/journal.pone.0051958] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 11/08/2012] [Indexed: 12/17/2022] Open
Abstract
Antibody-mediated imaging of amyloid β (Aβ) in Alzheimer's disease (AD) offers a promising strategy to detect and monitor specific Aβ species, such as oligomers, that have important pathological and therapeutic relevance. The major current limitation of antibodies as a diagnostic and imaging device is poor blood-brain-barrier permeability. A classical anti-Aβ antibody, 6E10, is modified with 10 kDa polyethylene glycol (PEG) and a positron emitting isotope, Copper-64 (t½ = 12.7 h), and intravenously delivered to the TgCRND8 mouse model of Alzheimer's disease. Modification of 6E10 with PEG (6E10-PEG) increases accumulation of 6E10 in brain tissue in both TgCRND8 and wild type control animals. 6E10-PEG differentiates TgCRND8 animals from wild type controls using positron emission tomography (PET) and provides a framework for using antibodies to detect pathology using non-invasive medical imaging techniques.
Collapse
|
47
|
Ferreira A. Calpain dysregulation in Alzheimer's disease. ISRN BIOCHEMISTRY 2012; 2012:728571. [PMID: 25969760 PMCID: PMC4393001 DOI: 10.5402/2012/728571] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 09/12/2012] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is characterized by the presence of senile plaques and neurofibrillary tangles in the neocortex and hippocampus of AD patients. In addition, a marked decrease in synaptic contacts has been detected in these affected brain areas. Due to its prevalence in the aging population, this disease has been the focus of numerous studies. The data obtained from those studies suggest that the mechanisms leading to the formation of the hallmark lesions of AD might be linked. One of such mechanisms seems to be the dysregulation of calcium homeostasis that results in the abnormal activation of calpains. Calpains are a family of Ca(2+)-dependent cysteine proteases that play a key role in multiple cell functions including cell development, differentiation and proliferation, axonal guidance, growth cone motility, and cell death, among others. In this paper, we briefly reviewed data on the structure of these proteases and their regulation under normal conditions. We also summarized data underscoring the participation of calpains in the neurodegenerative mechanisms associated with AD.
Collapse
Affiliation(s)
- Adriana Ferreira
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 8-140, Chicago, IL 60611, USA
| |
Collapse
|