1
|
Fan Y, Yang Y, Lin K, Zhou X, Li Y, Lin Q. The effect of AQP4 on tau protein aggregation in neurodegeneration and persistent neuroinflammation after cerebral microinfarcts. Open Med (Wars) 2023; 18:20230800. [PMID: 37873537 PMCID: PMC10590608 DOI: 10.1515/med-2023-0800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 10/25/2023] Open
Abstract
This study aimed to investigate the effect of aquaporin-4 (AQP4) on tau protein aggregation in neurodegeneration and persistent neuroinflammation after cerebral microinfarcts. A model of diffuse ischemic brain injury was established, and adenovirus was injected stereotactically through the lateral ventricle of mice. The water content of the brain tissue was measured. The co-expression of glial fibrillary acidic protein (GFAP) and AQP4 and the aggregation of p-tau and neuronal marker were detected through immunofluorescence double staining. The expression of phosphorylated microtubule-associated protein tau (p-tau, Ser202/Thr205, Thr205, Ser396, Ser404), interleukin(IL)-6, IL-1β, tumor necrosis factor (TNF)-a, growth associated protein43 (GAP43), GFAP, and ionized calcium-binding adapter molecule 1 (Iba1) was detected through Western blot. It was found that the brain water content in the model group was increased and decreased after the AQP4 interference. Compared with the sham group, the expression of GFAP, p-tau, IL-1β, TNF-a, Iba1, and p-tau was increased in the model group (p < 0.05). Compared with the model group, the expression of p-tau, IL-6, IL-1β, TNF-a, GFAP, and Iba1 was decreased after AQP4 interference (p < 0.05). It is indicated that AQP4 positively regulates neurodegeneration and persistent neuroinflammation caused by tau protein aggregation after cerebral microinfarcts.
Collapse
Affiliation(s)
- Yong Fan
- Central Laboratory, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Yongkai Yang
- Department of Neurosurgery, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Kunzhe Lin
- Department of Neurosurgery, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Xiaohui Zhou
- Department of Neurosurgery, Affiliated Fuzhou First Hospital of Fujian Medical University, Fuzhou, Fujian, 350009, China
| | - Yongkun Li
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No. 134, East Street, Fuzhou, Fujian, 350001, China
| | - Qingqiang Lin
- College of Life Sciences, Fujian Normal University, Qishan Campus, No. 13 Science and Engineering Building, Fuzhou, Fujian, 350117, China
| |
Collapse
|
2
|
Lee D, Ryoo JE, Hong S, Kim HY, Kim Y. Carprofen alleviates Alzheimer-like phenotypes of 5XFAD transgenic mice by targeting the pathological hallmarks induced by amyloid-β aggregation. Sci Rep 2023; 13:10889. [PMID: 37407605 DOI: 10.1038/s41598-023-36167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by misfolding, oligomerization, and accumulation of amyloid-β (Aβ) peptides in the brain. Aβ monomers transform into Aβ oligomers, which are toxic species, inducing tau hyperphosphorylation and the downstream effects on microglia and astrocytes, triggering synaptic and cognitive dysfunctions. The oligomers then deposit into Aβ plaques, primarily composed of β-stranded fibrils, required for definitive AD diagnosis. As amyloid burden plays the pivotal role in AD pathogenesis, many efforts are devoted in preventing amyloidosis as a therapeutic approach to impede the disease progression. Here, we discovered carprofen, a non-steroidal anti-inflammatory drug, accelerates Aβ aggregating into fibrils and increases Aβ plaques when intraperitoneally injected to 5XFAD transgenic mouse model. However, the drug seems to alleviate the key Alzheimer-like phenotypes induced by Aβ aggregation as we found attenuated neuroinflammation, improved post-synaptic density expression, associated with synaptic plasticity, and decreased phosphorylated tau levels. Carprofen also rescued impaired working memory as we discovered improved spontaneous alternation performance through Y-maze test assessed with Aβ(1-42)-infused mouse model. Collectively, while carprofen accelerates the conversion of Aβ monomers into fibrils in vitro, the drug ameliorates the major pathological hallmarks of AD in vivo.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Ji Eun Ryoo
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Seungpyo Hong
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Yonsei Frontier Lab, Yonsei University, Seoul, 03722, Republic of Korea
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
| | - Hye Yun Kim
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea.
- Yonsei Frontier Lab, Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon, 21983, Republic of Korea.
| |
Collapse
|
3
|
Bello-Medina PC, González-Franco DA, Vargas-Rodríguez I, Díaz-Cintra S. Oxidative stress, the immune response, synaptic plasticity, and cognition in transgenic models of Alzheimer disease. Neurologia 2022; 37:682-690. [PMID: 31780319 DOI: 10.1016/j.nrl.2019.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/27/2019] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Worldwide, approximately 50 million people have dementia, with Alzheimer disease (AD) being the most common type, accounting for 60%-70% of cases. Given its high incidence, it is imperative to design studies to expand our knowledge about its onset and development, and to develop early diagnosis strategies and/or possible treatments. One methodological strategy is the use of transgenic mouse models for the study of the factors involved in AD aetiology, which include oxidative stress and the immune response. DEVELOPMENT We searched the PubMed, Scopus, and Google Scholar databases for original articles and reviews published between 2013 and 2019. In this review, we address two factors that have been studied independently, oxidative stress and the immune response, in transgenic models of AD, and discuss the relationship between these factors and their impact on the loss of synaptic and structural plasticity, resulting in cognitive impairment. CONCLUSION This review describes possible mechanisms by which oxidative stress and the immune response participate in the molecular, cellular, and behavioural effects of AD, observing a close relationship between these factors, which lead to cognitive impairment.
Collapse
Affiliation(s)
- P C Bello-Medina
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - D A González-Franco
- Facultad de Psicología, Universidad Latina de México, Celaya, Guanajuato, México
| | - I Vargas-Rodríguez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - S Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México.
| |
Collapse
|
4
|
Li JM, Yang FH, Chao MW, Tseng CY. Swimming exercise prevents hippocampal dendritic spine changes and memory loss caused by aging: An application of a new semi-automated spine analysis software. Mol Cell Neurosci 2022; 121:103755. [PMID: 35850447 DOI: 10.1016/j.mcn.2022.103755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/06/2022] [Accepted: 07/10/2022] [Indexed: 11/17/2022] Open
Abstract
Dendritic spines are small, ratchet-like protrusions on neuronal dendrites that form synapses for receiving neuronal messages. Dendritic spine morphology is associated with synapse function. If neurons degrade or are damaged, the spine morphology of neurons changes. Given that most commercially available spine analysis software is expensive and complex, this study investigated a semi-automated spine analysis software, CTSpine, and used previously published data to verify the accuracy of the analysis results of this software. We also applied CTSpine to understand whether aging causes alterations in the hippocampal spine morphology and whether physical exercise can impede dendritic spine changes in 20 male Sprague Dawley rats. The spines of pyramidal cells in the hippocampal Cornu Ammonis 1 (CA1) region in the aging group were more enriched in filopodium type pattern than those in the control group, whereas the spines of the exercised aging group showed a similar pattern to that of the control. No significant changes were observed in neuronal dendritic spines in other hippocampal regions. However, long-term hippocampal memory was considerably decreased in the aging group, which was reversed to some extent in the exercised aging group. CTSpine, a self-developed semi-automatic spine analysis software, showed results similar to those noted in published data and can be effectively applied to the study of dendritic patterns, including neurodevelopment and disease.
Collapse
Affiliation(s)
- Jun-Ming Li
- Psychiatry Department, Taoyuan Armed Forces General Hospital, No. 168, Zhongxing Rd., Longtan Dist, Taoyuan City 32551, Taiwan.
| | - Fu-Hua Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| | - Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| | - Chia-Yi Tseng
- Department of Biomedical Engineering, Chung Yuan Christian University, Zhongli District, Taoyuan 320, Taiwan.
| |
Collapse
|
5
|
He X, Wang X, Yang L, Yang Z, Yu W, Wang Y, Liu R, Chen M, Gao H. Intelligent lesion blood brain barrier targeting nano-missiles for Alzheimer’s disease treatment by anti-neuroinflammation and neuroprotection. Acta Pharm Sin B 2022; 12:1987-1999. [PMID: 35847512 PMCID: PMC9279705 DOI: 10.1016/j.apsb.2022.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/14/2021] [Accepted: 01/10/2022] [Indexed: 12/16/2022] Open
Affiliation(s)
- Xueqin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lianyi Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Wenqi Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yazhen Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rui Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Corresponding author.
| |
Collapse
|
6
|
Abdel-Aal RA, Hussein OA, Elsaady RG, Abdelzaher LA. Naproxen as a potential candidate for promoting rivastigmine anti-Alzheimer activity against aluminum chloride-prompted Alzheimer's-like disease in rats; neurogenesis and apoptosis modulation as a possible underlying mechanism. Eur J Pharmacol 2022; 915:174695. [PMID: 34914971 DOI: 10.1016/j.ejphar.2021.174695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIM Alzheimer's disease (AD) is one of the leading causes of dependence and disability among the elderly worldwide. The traditional anti-Alzheimer medication, rivastigmine, one of the cholinesterase inhibitors (ChEIs), fails to achieve a definitive cure. We tested the hypothesis that naproxen administration to the rivastigmine-treated aluminum chloride (AlCl3) Alzheimer's rat model could provide an additive neuroprotective effect compared to rivastigmine alone. MATERIALS AND METHODS The studied groups were control (Cont), AlCl3 treated (Al), rivastigmine treated (RIVA), naproxen treated (Napro), and combined rivastigmine and naproxen treated (RIVA + Napro). Rats' memory, spatial learning, and cognitive behavior were assessed followed by evaluation of hippocampal acetylcholinesterase (AChE) activity. Hippocampal and cerebellar histopathology were thoroughly examined. Activated caspase-3 and the neuroepithelial stem cells marker; nestin expressions were immunohistochemically assayed. RESULTS AD rats displayed significantly impaired memory and cognitive function, augmented hippocampal AChE activity; massive neurodegeneration associated with enhanced astrogliosis, apoptosis, and impaired neurogenesis. Except for the enhancement of neurogenesis and suppression of apoptosis, the combination therapy had no additional neuroprotective benefit over rivastigmine-only therapy. CONCLUSION Naproxen's efficacy was established by its ability to function at the cellular level, improved neurogenesis, and decreased, apoptosis without having an additional mitigating impact on cognitive impairment in rivastigmine-treated AD rats.
Collapse
Affiliation(s)
- Raafat A Abdel-Aal
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ola A Hussein
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Reham G Elsaady
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
7
|
Onaolapo OJ, Olofinnade AT, Ojo FO, Onaolapo AY. Neuroinflammation and Oxidative Stress in Alzheimer's Disease; Can Nutraceuticals and Functional Foods Come to the Rescue? Antiinflamm Antiallergy Agents Med Chem 2022; 21:75-89. [PMID: 36043770 DOI: 10.2174/1871523021666220815151559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD), the most prevalent form of age-related dementia, is typified by progressive memory loss and spatial awareness with personality changes. The increasing socioeconomic burden associated with AD has made it a focus of extensive research. Ample scientific evidence supports the role of neuroinflammation and oxidative stress in AD pathophysiology, and there is increasing research into the possible role of anti-inflammatory and antioxidative agents as disease modifying therapies. While, the result of numerous preclinical studies has demonstrated the benefits of anti-inflammatory agents, these benefits however have not been replicated in clinical trials, necessitating a further search for more promising anti-inflammatory agents. Current understanding highlights the role of diet in the development of neuroinflammation and oxidative stress, as well as the importance of dietary interventions and lifestyle modifications in mitigating them. The current narrative review examines scientific literature for evidence of the roles (if any) of dietary components, nutraceuticals and functional foods in the prevention or management of AD. It also examines how diet/ dietary components could modulate oxidative stress/inflammatory mediators and pathways that are crucial to the pathogenesis and/or progression of AD.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Department of Pharmacology, Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Anthony T Olofinnade
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, College of Medicine, Lagos State University, Ikeja, Lagos State, Nigeria
| | - Folusho O Ojo
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Adejoke Y Onaolapo
- Department of Anatomy, Behavioural Neuroscience Unit, Neurobiology Subdivision, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
8
|
Han Y, Lu Y, Li X, Niu X, Chang AK, Yang Z, Li X, He X, Bi X. Novel organoselenides (NSAIDs-Se derivatives) protect against LPS-induced inflammation in microglia by targeting the NOX2/NLRP3 signaling pathway. Int Immunopharmacol 2021; 101:108377. [PMID: 34836795 DOI: 10.1016/j.intimp.2021.108377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/30/2021] [Accepted: 11/11/2021] [Indexed: 12/01/2022]
Abstract
Neuro-inflammation is an immune response of the central nervous system (CNS) to pathogens, and it is associated with a variety of neurodegenerative diseases. Microglial cells are the main category of macrophages in the CNS parenchyma, and they represent one of the most important cellular drivers and regulators of neuroinflammation. In this study, nine new organoselenium compounds based on the hybridization of nonsteroidal anti-inflammatory drugs (NSAIDs) skeleton and organoselenium motif (-SeCN and -SeCF3) were synthesized and their potential anti-neuroinflammatory effects were evaluated using LPS-induced BV2 mouse microglia. The cells were first treated with the organoselenium compounds and the extent of oxidative stress and inflammatory response of the cells was determined by measuring the levels of NO, ROS, IL-1β, and IL-18. Among the nine compounds, 1-39 and 1A-38 exhibited the most significant effect on oxidative stress and inflammatory response. Subsequent studies carried out with 1-39 and 1A-38 showed that both compounds could reduce the production of ROS in the cells, probably through down-regulating NOX2 and its downstream targets, including TXNIP (thioredoxin-interacting protein) and NLRP3 (NOD-like receptor protein 3). In addition, 1-39 and 1A-38 also suppressed the ability of the cells to secret IL-18 and IL-1β, which greatly dampened the response of the cells to LPS-induced inflammation. Our finding demonstrated that organoselenium compounds derived from NSAID might play an important role in the protection of brain microglia against inflammation-related neurodegenerative disease by potentially down-regulating the NOX2/NLRP3 signaling axis.
Collapse
Affiliation(s)
- Yunsu Han
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Yin Lu
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan 430056, China
| | - Xin Li
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Xiaoqi Niu
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Alan K Chang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou 325035, China
| | - Zhe Yang
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Xiaolong Li
- Shenzhen Fushan Biological Technology Co., Ltd, Kexing Science Park A1 1005, Nanshan Zone, Shenzhen 518057, China
| | - Xianran He
- Institute for Interdisciplinary Research, Jianghan University, Wuhan Economic and Technological Development Zone, Wuhan 430056, China.
| | - Xiuli Bi
- College of Life Science, Liaoning University, Shenyang 110036, China.
| |
Collapse
|
9
|
Kumar M, Bansal N. A Revisit to Etiopathogenesis and Therapeutic Strategies in Alzheimer's Disease. Curr Drug Targets 2021; 23:486-512. [PMID: 34792002 DOI: 10.2174/1389450122666211118125233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022]
Abstract
Dementia is a cluster of brain abnormalities that trigger progressive memory deficits and other cognitive abilities such as skills, language, or executive function. Alzheimer's disease (AD) is the foremost type of age-associated dementia that involves progressive neurodegeneration accompanied by profound cognitive deficits in advanced stages that severely hamper social or occupational abilities with or without the involvement of any other psychiatric condition. The last two decades witnessed a sharp increase (~123%) in mortality due to AD type dementia, typically owing to a very low disclosure rate (~45%) and hence, the prophylactic, as well as the therapeutic cure of AD, has been a huge challenge. Although understanding of AD pathogenesis has witnessed a remarkable growth (e.g., tauopathy, oxidative stress, lipid transport, glucose uptake, apoptosis, synaptic dysfunction, inflammation, and immune system), still a dearth of an effective therapeutic agent in the management of AD prompts the quest for newer pharmacological targets in the purview of its growing epidemiological status. Most of the current therapeutic strategies focus on modulation of a single target, e.g., inhibition of acetylcholinesterase, glutamate excitotoxicity (memantine), or nootropics (piracetam), even though AD is a multifaceted neurological disorder. There is an impedance urgency to find not only symptomatic but effective disease-modifying therapies. The present review focuses on the risk / protective factors and pathogenic mechanisms involved in AD. In addition to the existing symptomatic therapeutic approach, a diverse array of possible targets linked to pathogenic cascades have been re-investigated to envisage the pharmacotherapeutic strategies in AD.
Collapse
Affiliation(s)
- Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Nitin Bansal
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana 127021. India
| |
Collapse
|
10
|
Bello-Medina PC, González-Franco DA, Vargas-Rodríguez I, Díaz-Cintra S. Oxidative stress, the immune response, synaptic plasticity, and cognition in transgenic models of Alzheimer disease. NEUROLOGÍA (ENGLISH EDITION) 2021; 37:682-690. [PMID: 34509401 DOI: 10.1016/j.nrleng.2019.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 06/27/2019] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Worldwide, approximately 50 million people have dementia, with Alzheimer disease (AD) being the most common type, accounting for 60%-70% of cases. Given its high incidence, it is imperative to design studies to expand our knowledge about its onset and development, and to develop early diagnosis strategies and/or possible treatments. One methodological strategy is the use of transgenic mouse models for the study of the factors involved in AD aetiology, which include oxidative stress and the immune response. DEVELOPMENT We searched the PubMed, Scopus, and Google Scholar databases for original articles and reviews published between 2013 and 2019. In this review, we address 2 factors that have been studied independently, oxidative stress and the immune response, in transgenic models of AD, and discuss the relationship between these factors and their impact on the loss of synaptic and structural plasticity, resulting in cognitive impairment. CONCLUSION This review describes possible mechanisms by which oxidative stress and the immune response participate in the molecular, cellular, and behavioural effects of AD, observing a close relationship between these factors, which lead to cognitive impairment.
Collapse
Affiliation(s)
- P C Bello-Medina
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - D A González-Franco
- Facultad de Psicología, Universidad Latina de México, Celaya, Guanajuato, Mexico
| | - I Vargas-Rodríguez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico
| | - S Díaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, Mexico.
| |
Collapse
|
11
|
Mathis SP, Bodduluri SR, Haribabu B. Interrelationship between the 5-lipoxygenase pathway and microbial dysbiosis in the progression of Alzheimer's disease. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158982. [PMID: 34062254 PMCID: PMC11522975 DOI: 10.1016/j.bbalip.2021.158982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder involving neurofibrillary tangles and amyloid plaques. The tau phosphorylation responsible for neurofibrillary tangles and amyloid deposition which causes plaques are both accelerated through the activity of 5-lipoxygenase (5-LO). In addition to these pathological pathways, 5-LO has also been linked to the neuro-inflammation associated with disease progression as well as to dysbiosis in the gut. Interestingly, gut dysbiosis itself has been correlated to AD development. Not only do gut metabolites have direct effects on the brain, but pro-inflammatory mediators such as LPS, BMAA and bacterial amyloids produced in the gut due to dysbiosis reach the brain causing increased neuro-inflammation. While microbial dysbiosis and 5-LO exert detrimental effects in the brain, the cause/effect relationship between these factors remain unknown. These issues may be addressed using mouse models of AD in the context of different knockout mice in the 5-LO pathway in specific pathogen-free, germ-free as well as gnotobiotic conditions.
Collapse
Affiliation(s)
- Steven P Mathis
- Department of Microbiology and Immunology, James Graham Brown Cancer Center and Center for Microbiomics, Inflammation and Pathogenicity, Louisville, KY 40202, United States of America; University of Louisville Health Sciences Center, Louisville, KY 40202, United States of America
| | - Sobha R Bodduluri
- Department of Microbiology and Immunology, James Graham Brown Cancer Center and Center for Microbiomics, Inflammation and Pathogenicity, Louisville, KY 40202, United States of America; University of Louisville Health Sciences Center, Louisville, KY 40202, United States of America
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, James Graham Brown Cancer Center and Center for Microbiomics, Inflammation and Pathogenicity, Louisville, KY 40202, United States of America; University of Louisville Health Sciences Center, Louisville, KY 40202, United States of America.
| |
Collapse
|
12
|
Zhang X, Chau LY, Chan HW, Weng J, Wong KW, Chow SF, Chow AHL. Physical stability and in vivo brain delivery of polymeric ibuprofen nanoparticles fabricated by flash nanoprecipitation. Int J Pharm 2021; 598:120224. [PMID: 33486028 DOI: 10.1016/j.ijpharm.2021.120224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/06/2020] [Accepted: 12/24/2020] [Indexed: 01/16/2023]
Abstract
Ibuprofen (IBP), a common non-steroidal anti-inflammatory drug (NSAID) with a log P of 3.51, has been shown to possess potential benefit in the treatment of Alzheimer's disease. However, the bioavailability of IBP to the brain is poor, which can be linked to its extensive binding to plasma proteins in the blood. This study aimed to evaluate the nanoparticle production of IBP by flash nanoprecipitation (FNP) technology, and to determine whether the nanoparticles prepared by FNP could enhance the delivery of IBP into the brain. Polymeric IBP nanoparticles were prepared with poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) diblock copolymer as stabilizer under optimized conditions using a four-stream multi-inlet vortex mixer (MIVM). The optimized nanoparticles displayed a mean particle size of around 50 nm, polydispersity index of around 0.2, drug loading of up to 30% and physical stability of up to 34 days. In-depth surface characterization using zeta potential measurement, atomic force microscopy (AFM) and X-ray photoelectron spectroscopy (XPS) showed that the surfaces of these nanoparticles were covered with the hydrophilic PEG groups from the diblock copolymer. In vivo brain uptake study of the IBP nanoparticles indicated that the particles, when coated with polysorbate 80, displayed an enhanced brain uptake. However, the extent of brain uptake enhancement appeared limited, possibly due to a rapid release of IBP from the nanoparticles into the blood stream following intravenous administration.
Collapse
Affiliation(s)
- Xinran Zhang
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Li Yin Chau
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | - Ho Wan Chan
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Jingwen Weng
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Ka Wai Wong
- Genvida (HK) Company Limited, Hong Kong Special Administrative Region
| | - Shing Fung Chow
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region.
| | - Albert Hee Lum Chow
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region.
| |
Collapse
|
13
|
Sohn E, Kim YJ, Jeong SJ. Korean traditional herbal formula Soshiho-tang attenuates memory impairment and neuronal damage in mice with amyloid-beta-induced Alzheimer's disease. Integr Med Res 2021; 10:100723. [PMID: 33898246 PMCID: PMC8059063 DOI: 10.1016/j.imr.2021.100723] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/05/2021] [Accepted: 03/17/2021] [Indexed: 11/10/2022] Open
Abstract
Background Soshiho-tang (SST), also known as Xiaochaihu-tang in China and Sho-saiko-to in Japan, is an Oriental herbal formula traditionally used to treat febrile diseases. Recently, several in vitro and in vivo studies have reported the anti-cancer, anti-liver disease, and anti-inflammatory activities of SST. However, there is little evidence of its effects on neurological diseases. We previously reported the inhibitory effects of SST on in vitro acetylcholinesterase (AChE) activation and amyloid-β (Aβ) aggregation, which are crucial hallmarks of Alzheimer's disease (AD). In the present study, we report that SST has preventive effects on memory impairment and neuronal cell changes in an Aβ-induced AD-like mouse model. Methods Male mice underwent injection of Aβ aggregates and administered SST (500, 1,000, or 2,000 mg/kg/day) for 20 days. Behavioral tests (passive avoidance task [PAT] and Morris water maze [MWM] test) were conducted. Lastly, brain sections were obtained from sacrificed mice for quantitative analysis. Results Intracerebroventricular (ICV) injection of Aβ aggregates significantly decreased the latency time in the PAT and MWM test compared to normal control. In contrast, SST administration markedly reversed the latency caused by Aβ injection. Additionally, our data revealed that SST-mediated improvements in memory impairment are related to its neuroprotective and anti-neuroinflammatory effects. On histological analysis, SST treatment protected neuronal loss and damage as well as microglial activation, and ameliorated amount of Aβ in brain of mouse model of AD. Conclusion Our findings suggest that SST may be a promising candidate for the development of novel drugs for AD.
Collapse
Affiliation(s)
- Eunjin Sohn
- Clinical Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Yu Jin Kim
- Clinical Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Soo-Jin Jeong
- Clinical Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| |
Collapse
|
14
|
Liu Z, Zhang B, Xia S, Fang L, Gou S. ROS-responsive and multifunctional anti-Alzheimer prodrugs: Tacrine-ibuprofen hybrids via a phenyl boronate linker. Eur J Med Chem 2020; 212:112997. [PMID: 33189440 DOI: 10.1016/j.ejmech.2020.112997] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Current drugs available in clinic for Alzheimer's disease (AD) treatment can only alleviate disease symptoms without clearly curing or delaying the process of AD. And some AD drugs failed in Phase III clinical trials are only focused on targeting amyloid-β (Aβ). Therefore, an alternative strategy in AD drug design is meaningful to be involved in the multiple pathogenic factors which can affect each other at multiple levels. Herein, we report a series of ROS-responsive prodrugs based on multi-target-directed ligands (MTDLs) approach, which can specifically release tacrine derivatives and ibuprofen under oxidation of ROS and show acetylcholinesterase (AChE)-inhibiting, neuron-protective and anti-inflammatory effects in extracellular or intracellular assays. Related biological study illustrated that compound 22 was able to permeate blood-brain-barrier (BBB) showing little hepatotoxicity in comparison to tacrine. Besides, 22 hinted a therapeutic clue in AD-treatment by regulating proinflammatory factors (IL-1β and TNF-α) and apoptosis related proteins (Bax, Bcl-2 and cleaved caspase-3). Further spatial memory assays in Aβ-induced AD model showed that 22 enhanced the ability of learning and memory. Our study proves that the strategy of ROS-responsive prodrugs has promise for AD treatments in future and offers a way for AD drug development.
Collapse
Affiliation(s)
- Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Bin Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Shengjin Xia
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Lei Fang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| | - Shaohua Gou
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing, 211189, China; Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
15
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer's Disease Neurodegeneration in Individuals with and without Down Syndrome. Int J Mol Sci 2020; 21:E6906. [PMID: 32962300 PMCID: PMC7555886 DOI: 10.3390/ijms21186906] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer's disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain;
| | | |
Collapse
|
17
|
Flores J, Noël A, Foveau B, Beauchet O, LeBlanc AC. Pre-symptomatic Caspase-1 inhibitor delays cognitive decline in a mouse model of Alzheimer disease and aging. Nat Commun 2020; 11:4571. [PMID: 32917871 PMCID: PMC7486940 DOI: 10.1038/s41467-020-18405-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/20/2020] [Indexed: 02/07/2023] Open
Abstract
Early therapeutic interventions are essential to prevent Alzheimer Disease (AD). The association of several inflammation-related genetic markers with AD and the early activation of pro-inflammatory pathways in AD suggest inflammation as a plausible therapeutic target. Inflammatory Caspase-1 has a significant impact on AD-like pathophysiology and Caspase-1 inhibitor, VX-765, reverses cognitive deficits in AD mouse models. Here, a one-month pre-symptomatic treatment of Swedish/Indiana mutant amyloid precursor protein (APPSw/Ind) J20 and wild-type mice with VX-765 delays both APPSw/Ind- and age-induced episodic and spatial memory deficits. VX-765 delays inflammation without considerably affecting soluble and aggregated amyloid beta peptide (Aβ) levels. Episodic memory scores correlate negatively with microglial activation. These results suggest that Caspase-1-mediated inflammation occurs early in the disease and raise hope that VX-765, a previously Food and Drug Administration-approved drug for human CNS clinical trials, may be a useful drug to prevent the onset of cognitive deficits and brain inflammation in AD.
Collapse
Affiliation(s)
- Joseph Flores
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Anastasia Noël
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Bénédicte Foveau
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Olivier Beauchet
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,Department of Medicine, Division of Geriatric Medicine, Sir Mortimer B. Davis - Jewish General Hospital, Montreal, Quebec, Canada.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Andréa C LeBlanc
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada. .,Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Mendonça LS, Nóbrega C, Tavino S, Brinkhaus M, Matos C, Tomé S, Moreira R, Henriques D, Kaspar BK, Pereira de Almeida L. Ibuprofen enhances synaptic function and neural progenitors proliferation markers and improves neuropathology and motor coordination in Machado-Joseph disease models. Hum Mol Genet 2020; 28:3691-3703. [PMID: 31127937 DOI: 10.1093/hmg/ddz097] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 01/09/2023] Open
Abstract
Machado-Joseph disease or spinocerebellar ataxia type 3 is an inherited neurodegenerative disease associated with an abnormal glutamine over-repetition within the ataxin-3 protein. This mutant ataxin-3 protein affects several cellular pathways, leading to neuroinflammation and neuronal death in specific brain regions resulting in severe clinical manifestations. Presently, there is no therapy able to modify the disease progression. Nevertheless, anti-inflammatory pharmacological intervention has been associated with positive outcomes in other neurodegenerative diseases. Thus, the present work aimed at investigating whether ibuprofen treatment would alleviate Machado-Joseph disease. We found that ibuprofen-treated mouse models presented a significant reduction in the neuroinflammation markers, namely Il1b and TNFa mRNA and IKB-α protein phosphorylation levels. Moreover, these mice exhibited neuronal preservation, cerebellar atrophy reduction, smaller mutant ataxin-3 inclusions and motor performance improvement. Additionally, neural cultures of Machado-Joseph disease patients' induced pluripotent stem cells-derived neural stem cells incubated with ibuprofen showed increased levels of neural progenitors proliferation and synaptic markers such as MSI1, NOTCH1 and SYP. These findings were further confirmed in ibuprofen-treated mice that display increased neural progenitor numbers (Ki67 positive) in the subventricular zone. Furthermore, interestingly, ibuprofen treatment enhanced neurite total length and synaptic function of human neurons. Therefore, our results indicate that ibuprofen reduces neuroinflammation and induces neuroprotection, alleviating Machado-Joseph disease-associated neuropathology and motor impairments. Thus, our findings demonstrate that ibuprofen treatment has the potential to be used as a neuroprotective therapeutic approach in Machado-Joseph disease.
Collapse
Affiliation(s)
- Liliana S Mendonça
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Clévio Nóbrega
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Silvia Tavino
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Maximilian Brinkhaus
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Carlos Matos
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra Tomé
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ricardo Moreira
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Daniel Henriques
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Ohio State University School of Medicine, Columbus, Ohio 43205, USA
| | - Luís Pereira de Almeida
- Vectors and Gene Therapy Group, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
19
|
Girigoswami A, Ramalakshmi M, Akhtar N, Metkar SK, Girigoswami K. ZnO Nanoflower petals mediated amyloid degradation - An in vitro electrokinetic potential approach. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:169-178. [DOI: 10.1016/j.msec.2019.03.086] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/25/2019] [Accepted: 03/24/2019] [Indexed: 10/27/2022]
|
20
|
Medrano-Jiménez E, Jiménez-Ferrer Carrillo I, Pedraza-Escalona M, Ramírez-Serrano CE, Álvarez-Arellano L, Cortés-Mendoza J, Herrera-Ruiz M, Jiménez-Ferrer E, Zamilpa A, Tortoriello J, Pedraza-Alva G, Pérez-Martínez L. Malva parviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer's disease by restoring microglial function via a PPAR-γ-dependent mechanism. J Neuroinflammation 2019; 16:143. [PMID: 31291963 PMCID: PMC6617588 DOI: 10.1186/s12974-019-1515-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a neuropathology strongly associated with the activation of inflammatory pathways. Accordingly, inflammation resulting from obesity exacerbates learning and memory deficits in humans and in animal models of AD. Consequently, the long-term use of non-steroidal anti-inflammatory agents diminishes the risk for developing AD, but the side effects produced by these drugs limit their prophylactic use. Thus, plants natural products have become an excellent option for modern therapeutics. Malva parviflora is a plant well known for its anti-inflammatory properties. Methods The present study was aimed to determine the anti-inflammatory potential of M. parviflora leaf hydroalcoholic extract (MpHE) on AD pathology in lean and obese transgenic 5XFAD mice, a model of familial AD. The inflammatory response and Amyloid β (Aβ) plaque load in lean and obese 5XFAD mice untreated or treated with MpHE was evaluated by immunolocalization (Iba-1 and GFAP) and RT-qPCR (TNF) assays and thioflavin-S staining, respectively. Spatial learning memory was assessed by the Morris Water Maze behavioral test. Microglia phagocytosis capacity was analyzed in vivo and by ex vivo and in vitro assays, and its activation by morphological changes (phalloidin staining) and expression of CD86, Mgl1, and TREM-2 by RT-qPCR. The mechanism triggered by the MpHE was characterized in microglia primary cultures and ex vivo assays by immunoblot (PPAR-γ) and RT-qPCR (CD36) and in vivo by flow cytometry, using GW9662 (PPAR-γ inhibitor) and pioglitazone (PPAR-γ agonist). The presence of bioactive compounds in the MpHE was determined by HPLC. Results MpHE efficiently reduced astrogliosis, the presence of insoluble Aβ peptides in the hippocampus and spatial learning impairments, of both, lean, and obese 5XFAD mice. This was accompanied by microglial cells accumulation around Aβ plaques in the cortex and the hippocampus and decreased expression of M1 inflammatory markers. Consistent with the fact that the MpHE rescued microglia phagocytic capacity via a PPAR-γ/CD36-dependent mechanism, the MpHE possess oleanolic acid and scopoletin as active phytochemicals. Conclusions M. parviflora suppresses neuroinflammation by inhibiting microglia pro-inflammatory M1 phenotype and promoting microglia phagocytosis. Therefore, M. parviflora phytochemicals represent an alternative to prevent cognitive impairment associated with a metabolic disorder as well as an effective prophylactic candidate for AD progression. Electronic supplementary material The online version of this article (10.1186/s12974-019-1515-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisa Medrano-Jiménez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Itzia Jiménez-Ferrer Carrillo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Martha Pedraza-Escalona
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Cristina E Ramírez-Serrano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Lourdes Álvarez-Arellano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México.,Present address: CONACYT-Hospital Infantil de México Federico Gómez, CP 06720, Ciudad de México, México
| | - Javier Cortés-Mendoza
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Jaime Tortoriello
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México.
| |
Collapse
|
21
|
Zhang C, Qin H, Zheng R, Wang Y, Yan T, Huan F, Han Y, Zhu W, Zhang L. A new approach for Alzheimer's disease treatment through P-gp regulation via ibuprofen. Pathol Res Pract 2018; 214:1765-1771. [PMID: 30139557 DOI: 10.1016/j.prp.2018.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
This study was aimed to investigate whether ibuprofen could alter the P-glycoprotein expression and function under Alzheimer's Disease condition and whether this alteration was induced by the inhibition of inflammatory reaction. APP/PS1 mice were used as AD model mice and ibuprofen-treated AD mice were given ibuprofen for 5 months. Then, Abcb1a/1b mRNA levels and P-gp expression were evaluated by qRT-PCR and western blot. Abcb1 mRNA levels were significantly reduced in AD mice compared to control mice, and it could be restored by ibuprofen treatment. Meanwhile, P-gp expression result showed a similar trend. Aβ plaques in cerebral cortices and hippocampus were investigated via immunohistochemical, and the results revealed that Aβ plaques were reduced in ibuprofen-treated AD mice compared with the AD mice, indicated that P-gp function may be recovered by ibuprofen treatment. qRT-PCR and ELISA were used to determined TNF-α, IL-1β, IL-6 and NF-κB levels. The results demonstrated that TNF-α, IL-1β mRNA levels and NF-κB expression were all significantly upregulated in AD mice in comparison with the control mice, and ibuprofen treatment could suppress the increase of inflammatory factors. In conclusion, the P-gp expression and function were suppressed in AD condition by activating inflammatory reaction, and then causing the Aβ efflux decreased. However, upregulating P-gp could increase the Aβ efflux in further to treat AD via inhibiting the inflammatory factors expression.
Collapse
Affiliation(s)
- Chengxiang Zhang
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Heng Qin
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Rui Zheng
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Yubang Wang
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Ting Yan
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Fei Huan
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Yang Han
- The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Weicheng Zhu
- The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| | - Lulu Zhang
- The Key Laboratory of Modern Toxicology of Ministry of Education, Department of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; The Safety Assessment and Research Center for Drugs, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
22
|
Abstract
Elevated levels of cyclooxygenase-2 (COX-2) and prostaglandins (PGs) are involved in the pathogenesis of Alzheimer's disease (AD), which is characterized by the accumulation of β-amyloid protein (Aβ) and tau hyperphosphorylation. However, the gaps in our knowledge of the roles of COX-2 and PGs in AD have not been filled. Here, we summarized the literature showing that COX-2 dysregulation obviously influences abnormal cleavage of β-amyloid precursor protein, aggregation and deposition of Aβ in β-amyloid plaques and the inclusion of phosphorylated tau in neurofibrillary tangles. Neuroinflammation, oxidative stress, synaptic plasticity, neurotoxicity, autophagy, and apoptosis have been assessed to elucidate the mechanisms of COX-2 regulation of AD. Notably, an imbalance of these factors ultimately produces cognitive decline. The current review substantiates our understanding of the mechanisms of COX-2-induced AD and establishes foundations for the design of feasible therapeutic strategies to treat AD.-Guan, P.-P., Wang, P. Integrated communications between cyclooxygenase-2 and Alzheimer's disease.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
23
|
Aguilar BJ, Zhu Y, Lu Q. Rho GTPases as therapeutic targets in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2017; 9:97. [PMID: 29246246 PMCID: PMC5732365 DOI: 10.1186/s13195-017-0320-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
The progress we have made in understanding Alzheimer’s disease (AD) pathogenesis has led to the identification of several novel pathways and potential therapeutic targets. Rho GTPases have been implicated as critical components in AD pathogenesis, but their various functions and interactions make understanding their complex signaling challenging to study. Recent advancements in both the field of AD and Rho GTPase drug development provide novel tools for the elucidation of Rho GTPases as a viable target for AD. Herein, we summarize the fluctuating activity of Rho GTPases in various stages of AD pathogenesis and in several in vitro and in vivo AD models. We also review the current pharmacological tools such as NSAIDs, RhoA/ROCK, Rac1, and Cdc42 inhibitors used to target Rho GTPases and their use in AD-related studies. Finally, we summarize the behavioral modifications following Rho GTPase modulation in several AD mouse models. As key regulators of several AD-related signals, Rho GTPases have been studied as targets in AD. However, a consensus has yet to be reached regarding the stage at which targeting Rho GTPases would be the most beneficial. The studies discussed herein emphasize the critical role of Rho GTPases and the benefits of their modulation in AD.
Collapse
Affiliation(s)
- Byron J Aguilar
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA.
| | - Yi Zhu
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA
| | - Qun Lu
- Department of Anatomy and Cell Biology, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA. .,The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Diseases Research, Brody School of Medicine at East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
24
|
Bansal R, Singh R. Exploring the potential of natural and synthetic neuroprotective steroids against neurodegenerative disorders: A literature review. Med Res Rev 2017; 38:1126-1158. [PMID: 28697282 DOI: 10.1002/med.21458] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Neurodegeneration is a complex process, which leads to progressive brain damage due to loss of neurons. Despite exhaustive research, the cause of neuronal loss in various degenerative disorders is not entirely understood. Neuroprotective steroids constitute an important line of attack, which could play a major role against the common mechanisms associated with various neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Natural endogenous steroids induce the neuroprotection by protecting the nerve cells from neuronal injury through multiple mechanisms, therefore the structural modifications of the endogenous steroids could be helpful in the generation of new therapeutically useful neuroprotective agents. The review article will keep the readers apprised of the detailed description of natural as well as synthetic neuroprotective steroids from the medicinal chemistry point of view, which would be helpful in drug discovery efforts aimed toward neurodegenerative diseases.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ranjit Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
25
|
Cornec AS, Monti L, Kovalevich J, Makani V, James MJ, Vijayendran KG, Oukoloff K, Yao Y, Lee VMY, Trojanowski JQ, Smith AB, Brunden KR, Ballatore C. Multitargeted Imidazoles: Potential Therapeutic Leads for Alzheimer's and Other Neurodegenerative Diseases. J Med Chem 2017; 60:5120-5145. [PMID: 28530811 PMCID: PMC5483893 DOI: 10.1021/acs.jmedchem.7b00475] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Alzheimer’s
disease (AD) is a complex, multifactorial disease in which different
neuropathological mechanisms are likely involved, including those
associated with pathological tau and Aβ species as well as neuroinflammation.
In this context, the development of single multitargeted therapeutics
directed against two or more disease mechanisms could be advantageous.
Starting from a series of 1,5-diarylimidazoles with microtubule (MT)-stabilizing
activity and structural similarities with known NSAIDs, we conducted
structure–activity relationship studies that led to the identification
of multitargeted prototypes with activities as MT-stabilizing agents
and/or inhibitors of the cyclooxygenase (COX) and 5-lipoxygenase (5-LOX)
pathways. Several examples are brain-penetrant and exhibit balanced
multitargeted in vitro activity in the low μM range. As brain-penetrant
MT-stabilizing agents have proven effective against tau-mediated neurodegeneration
in animal models, and because COX- and 5-LOX-derived eicosanoids are
thought to contribute to Aβ plaque deposition, these 1,5-diarylimidazoles
provide tools to explore novel multitargeted strategies for AD and
other neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne-Sophie Cornec
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania , 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Ludovica Monti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jane Kovalevich
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Vishruti Makani
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Michael J James
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Krishna G Vijayendran
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania , 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Killian Oukoloff
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yuemang Yao
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Amos B Smith
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania , 231 South 34th Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Kurt R Brunden
- Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania , 3600 Spruce Street, Philadelphia, Pennsylvania 19104-6323, United States
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego , 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
26
|
Sekiyama K, Takamatsu Y, Koike W, Waragai M, Takenouchi T, Sugama S, Hashimoto M. Insight into the Dissociation of Behavior from Histology in Synucleinopathies and in Related Neurodegenerative Diseases. J Alzheimers Dis 2017; 52:831-41. [PMID: 27031478 DOI: 10.3233/jad-151015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recent clinical trials using immunization approaches against Alzheimer's disease (AD) have failed to demonstrate improved cognitive functions in patients, despite potent suppression in the formation of both senile plaques and other amyloid-β deposits in postmortem brains. Similarly, we observed that treatment with ibuprofen, a non-steroidal anti-inflammatory drug, was effective in improving the histopathology, such as reducing both protein aggregation and glial activation, in the brains of transgenic mice expressing dementia with Lewy bodies-linked P123H β-synuclein. In contrast, only a small improvement in cognitive functions was observed in these mice. Collectively, it is predicted that histology does not correlate with behavior that is resilient and resistant to therapeutic stimuli. Notably, such a 'discrepancy between histology and behavior' is reminiscent of AD-like pathologies and incidental Lewy bodies, which are frequently encountered in postmortem brains of the elderly who had been asymptomatic for memory loss and Parkinsonism during their lives. We suggest that 'the discrepancy between histology and behavior' may be a universal feature that is associated with various aspects of neurodegenerative diseases. Furthermore, given that the cognitive reserve is specifically observed in human brains, human behavior may be evolutionally distinct from that in other animals, thus, contributing to the differential efficiency of therapy between human and lower animals, an important issue in the therapy of neurodegenerative diseases. Overall, it is important to better understand 'the discrepancy between histology and behavior' in the mechanism of neurodegeneration for the development of effective therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Kazunari Sekiyama
- Tokyo Metropolitan Institute of Medical Sciences, Setagaya-ku, Tokyo, Japan
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Sciences, Setagaya-ku, Tokyo, Japan
| | - Wakako Koike
- Tokyo Metropolitan Institute of Medical Sciences, Setagaya-ku, Tokyo, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Sciences, Setagaya-ku, Tokyo, Japan
| | - Takato Takenouchi
- Division of Animal Sciences, National Institute of Agrobiological Sciences, Tsukuba, Ibaraki, Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Sciences, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
27
|
Márquez Loza A, Elias V, Wong CP, Ho E, Bermudez M, Magnusson KR. Effects of ibuprofen on cognition and NMDA receptor subunit expression across aging. Neuroscience 2017; 344:276-292. [PMID: 28057539 DOI: 10.1016/j.neuroscience.2016.12.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/13/2016] [Accepted: 12/22/2016] [Indexed: 11/28/2022]
Abstract
Age-related declines in long- and short-term memory show relationships to decreases in N-methyl-d-aspartate (NMDA) receptor expression, which may involve inflammation. This study was designed to determine effects of an anti-inflammatory drug, ibuprofen, on cognitive function and NMDA receptor expression across aging. Male C57BL/6 mice (ages 5, 14, 20, and 26months) were fed ibuprofen (375ppm) in NIH31 diet or diet alone for 6weeks prior to testing. Behavioral testing using the Morris water maze showed that older mice performed significantly worse than younger in spatial long-term memory, reversal, and short-term memory tasks. Ibuprofen enhanced overall performance in the short-term memory task, but this appeared to be more related to improved executive function than memory. Ibuprofen induced significant decreases over all ages in the mRNA densities for GluN2B subunit, all GluN1 splice variants, and GluN1-1 splice forms in the frontal cortex and in protein expression of GluN2A, GluN2B and GluN1 C2' cassettes in the hippocampus. GluN1-3 splice form mRNA and C2' cassette protein were significantly increased across ages in frontal lobes of ibuprofen-treated mice. Ibuprofen did not alter expression of pro-inflammatory cytokines IL-1β and TNFα, but did reduce the area of reactive astrocyte immunostaining in frontal cortex of aged mice. Enhancement in executive function showed a relationship to increased GluN1-3 mRNA and decreased gliosis. These findings suggest that inflammation may play a role in executive function declines in aged animals, but other effects of ibuprofen on NMDA receptors appeared to be unrelated to aging or inflammation.
Collapse
Affiliation(s)
- Alejandra Márquez Loza
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Valerie Elias
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Carmen P Wong
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA; School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Michelle Bermudez
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Kathy R Magnusson
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
28
|
Ardura-Fabregat A, Boddeke EWGM, Boza-Serrano A, Brioschi S, Castro-Gomez S, Ceyzériat K, Dansokho C, Dierkes T, Gelders G, Heneka MT, Hoeijmakers L, Hoffmann A, Iaccarino L, Jahnert S, Kuhbandner K, Landreth G, Lonnemann N, Löschmann PA, McManus RM, Paulus A, Reemst K, Sanchez-Caro JM, Tiberi A, Van der Perren A, Vautheny A, Venegas C, Webers A, Weydt P, Wijasa TS, Xiang X, Yang Y. Targeting Neuroinflammation to Treat Alzheimer's Disease. CNS Drugs 2017; 31:1057-1082. [PMID: 29260466 PMCID: PMC5747579 DOI: 10.1007/s40263-017-0483-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Over the past few decades, research on Alzheimer's disease (AD) has focused on pathomechanisms linked to two of the major pathological hallmarks of extracellular deposition of beta-amyloid peptides and intra-neuronal formation of neurofibrils. Recently, a third disease component, the neuroinflammatory reaction mediated by cerebral innate immune cells, has entered the spotlight, prompted by findings from genetic, pre-clinical, and clinical studies. Various proteins that arise during neurodegeneration, including beta-amyloid, tau, heat shock proteins, and chromogranin, among others, act as danger-associated molecular patterns, that-upon engagement of pattern recognition receptors-induce inflammatory signaling pathways and ultimately lead to the production and release of immune mediators. These may have beneficial effects but ultimately compromise neuronal function and cause cell death. The current review, assembled by participants of the Chiclana Summer School on Neuroinflammation 2016, provides an overview of our current understanding of AD-related immune processes. We describe the principal cellular and molecular players in inflammation as they pertain to AD, examine modifying factors, and discuss potential future therapeutic targets.
Collapse
Affiliation(s)
- A. Ardura-Fabregat
- grid.5963.9Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - E. W. G. M. Boddeke
- 0000 0004 0407 1981grid.4830.fDepartment of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A. Boza-Serrano
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - S. Brioschi
- grid.5963.9Department of Psychiatry and Psychotherapy, Medical Center University of Freiburg, Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - S. Castro-Gomez
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Ceyzériat
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Dansokho
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - T. Dierkes
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dBiomedical Centre, Institute of Innate Immunity, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - G. Gelders
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Michael T. Heneka
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany ,0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - L. Hoeijmakers
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - A. Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - L. Iaccarino
- grid.15496.3fVita-Salute San Raffaele University, Milan, Italy ,0000000417581884grid.18887.3eIn Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S. Jahnert
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - K. Kuhbandner
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - G. Landreth
- 0000 0001 2287 3919grid.257413.6Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - N. Lonnemann
- 0000 0001 1090 0254grid.6738.aDepartment of Cellular Neurobiology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - R. M. McManus
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Paulus
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| | - K. Reemst
- 0000000084992262grid.7177.6Center for Neuroscience (SILS-CNS), Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - J. M. Sanchez-Caro
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - A. Tiberi
- grid.6093.cBio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - A. Van der Perren
- 0000 0001 0668 7884grid.5596.fDepartment of Neurosciences, Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - A. Vautheny
- grid.457334.2Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, MIRCen, 92260 Fontenay-aux-Roses, France ,0000 0001 2171 2558grid.5842.bNeurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, UMR 9199, F-92260 Fontenay-aux-Roses, France
| | - C. Venegas
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - A. Webers
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - P. Weydt
- 0000 0000 8786 803Xgrid.15090.3dDepartment of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn Medical Center, Sigmund-Freud Str. 25, 53127 Bonn, Germany
| | - T. S. Wijasa
- 0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), Sigmund Freud Str. 27, 53127 Bonn, Germany
| | - X. Xiang
- 0000 0004 1936 973Xgrid.5252.0Biomedical Center (BMC), Biochemistry, Ludwig-Maximilians-University Munich, 81377 Munich, Germany ,0000 0004 1936 973Xgrid.5252.0Graduate School of Systemic Neuroscience, Ludwig-Maximilians-University, Munich, 82152 Munich, Germany
| | - Y. Yang
- 0000 0001 0930 2361grid.4514.4Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences, Biomedical Centrum (BMC), Lund University, Lund, Sweden
| |
Collapse
|
29
|
Vallée A, Lecarpentier Y. Alzheimer Disease: Crosstalk between the Canonical Wnt/Beta-Catenin Pathway and PPARs Alpha and Gamma. Front Neurosci 2016; 10:459. [PMID: 27807401 PMCID: PMC5069291 DOI: 10.3389/fnins.2016.00459] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
The molecular mechanisms underlying the pathophysiology of Alzheimer's disease (AD) are still not fully understood. In AD, Wnt/beta-catenin signaling has been shown to be downregulated while the peroxisome proliferator-activated receptor (PPAR) gamma (mARN and protein) is upregulated. Certain neurodegenerative diseases share the same Wnt/beta-catenin/PPAR gamma profile, such as bipolar disorder and schizophrenia. Conversely, other NDs share an opposite profile, such as amyotrophic lateral sclerosis, Parkinson's disease, Huntington's disease, multiple sclerosis, and Friedreich's ataxia. AD is characterized by the deposition of extracellular Abeta plaques and the formation of intracellular neurofibrillary tangles in the central nervous system (CNS). Activation of Wnt signaling or inhibition of both glycogen synthase kinase-3beta and Dickkopf 1, two key negative regulators of the canonical Wnt pathway, are able to protect against Abeta neurotoxicity and to ameliorate cognitive performance in AD patients. Although PPAR gamma is upregulated in AD patients, and despite the fact that it has been shown that the PPAR gamma and Wnt/beta catenin pathway systems work in an opposite manner, PPAR gamma agonists diminish learning and memory deficits, decrease Abeta activation of microglia, and prevent hippocampal and cortical neurons from dying. These beneficial effects observed in AD transgenic mice and patients might be partially due to the anti-inflammatory properties of PPAR gamma agonists. Moreover, activation of PPAR alpha upregulates transcription of the alpha-secretase gene and represents a new therapeutic treatment for AD. This review focuses largely on the behavior of two opposing pathways in AD, namely Wnt/beta-catenin signaling and PPAR gamma. It is hoped that this approach may help to develop novel AD therapeutic strategies integrating PPAR alpha signaling.
Collapse
Affiliation(s)
- Alexandre Vallée
- CHU Amiens Picardie, Université Picardie Jules VerneAmiens, France
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of PoitiersPoitiers, France
- AP-HP, Epidemiology and Clinical Research Department, University Hospital Bichat-Claude BernardParis, France
| | | |
Collapse
|
30
|
Nagae T, Araki K, Shimoda Y, Sue LI, Beach TG, Konishi Y. Cytokines and Cytokine Receptors Involved in the Pathogenesis of Alzheimer's Disease. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2016; 7:441. [PMID: 27895978 PMCID: PMC5123596 DOI: 10.4172/2155-9899.1000441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inflammatory mechanisms are implicated in the pathology of Alzheimer's disease (AD). However, it is unclear whether inflammatory alterations are a cause or consequence of neurodegeneration leading to dementia. Clarifying this issue would provide valuable insight into the early diagnosis and therapeutic management of AD. To address this, we compared the mRNA expression profiles of cytokines in the brains of AD patients with "non-demented individuals with AD pathology" and non-demented healthy control (ND) individuals. "Non-demented individuals with AD pathology" are referred to as high pathology control (HPC) individuals that are considered an intermediate subset between AD and ND. HPC represents a transition between normal aging and early stage of AD, and therefore, is useful for determining whether neuroinflammation is a cause or consequence of AD pathology. We observed that immunological conditions that produce cytokines in the HPC brain were more representative of ND than AD. To validate these result, we investigated the expression of inflammatory mediators at the protein level in postmortem brain tissues. We examined the protein expression of tumor necrosis factor (TNF)α and its receptors (TNFRs) in the brains of AD, HPC, and ND individuals. We found differences in soluble TNFα and TNFRs expression between AD and ND groups and between AD and HPC groups. Expression in the temporal cortex was lower in the AD brains than HPC and ND. Our findings indicate that alterations in immunological conditions involving TNFR-mediated signaling are not the primary events initiating AD pathology, such as amyloid plaques and tangle formation. These may be early events occurring along with synaptic and neuronal changes or later events caused by these changes. In this review, we emphasize that elucidating the temporal expression of TNFα signaling molecules during AD is important to understand the selective tuning of these pathways required to develop effective therapeutic strategies for AD.
Collapse
Affiliation(s)
- Tomone Nagae
- Department of Clinical Research, National Tottori Medical Center, Tottori 689-0203, Japan
| | - Kiho Araki
- Department of Clinical Research, National Tottori Medical Center, Tottori 689-0203, Japan
| | - Yuki Shimoda
- Department of Clinical Research, National Tottori Medical Center, Tottori 689-0203, Japan
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Yoshihiro Konishi
- Department of Clinical Research, National Tottori Medical Center, Tottori 689-0203, Japan
| |
Collapse
|
31
|
Karthick C, Periyasamy S, Jayachandran KS, Anusuyadevi M. Intrahippocampal Administration of Ibotenic Acid Induced Cholinergic Dysfunction via NR2A/NR2B Expression: Implications of Resveratrol against Alzheimer Disease Pathophysiology. Front Mol Neurosci 2016; 9:28. [PMID: 27199654 PMCID: PMC4844917 DOI: 10.3389/fnmol.2016.00028] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/01/2016] [Indexed: 01/27/2023] Open
Abstract
Although several drugs revealed moderate amelioration of symptoms, none of them have sufficient potency to prevent or reverse the progression toward Alzheimer's disease (AD) pathology. Resveratrol (RSV), a polyphenolic compound has shown an outstanding therapeutic effect on a broad spectrum of diseases like age-associated neurodegeneration, inflammation etc. The present study was thus conducted to assess the therapeutic efficacy of RSV in ameliorating the deleterious effects of Ibotenic acid (IBO) in male Wistar rats. Stereotactic intrahippocampal administration of IBO (5 μg/μl) lesioned rats impairs cholinergic transmission, learning and memory performance that is rather related to AD and thus chosen as a suitable model to understand the drug efficacy in preventing AD pathophysiology. Since IBO is an agonist of glutamate, it is expected to exhibit an excitotoxic effect by altering glutamatergic receptors like NMDA receptor. The current study displayed significant alterations in the mRNA expression of NR2A and NR2B subunits of NMDA receptors, and further it is surprising to note that cholinergic receptors decreased in expression particularly α7-nAChR with increased m1AChR. RSV administration (20 mg/kg body weight, i.p.) significantly reduced these changes in IBO induced rats. Glutamatergic and cholinergic receptor alterations were associated with significant changes in the behavioral parameters of rats induced by IBO. While RSV improved spatial learning performance, attenuated immobility, and improvised open field activity in IBO induced rats. NR2B activation in the present study might mediate cell death through oxidative stress that form the basis of abnormal behavioral pattern in IBO induced rats. Interestingly, RSV that could efficiently encounter oxidative stress have significantly decreased stress markers viz., nitrite, PCO, and MDA levels by enhancing antioxidant status. Histopathological analysis displayed significant reduction in the hippocampal pyramidal layer thickness and live neurons in IBO induced rats, with slight pathological changes in the entorhinal cortex (EC) of rat brain, which was prevented on RSV administration. Our study thus concludes that RSV administration significantly ameliorated the deleterious effects in the IBO lesioned rat model for AD by alleviating cholinergic pathways, reducing oxidative stress and thereby improving spatial memory.
Collapse
Affiliation(s)
- Chennakesavan Karthick
- Molecular Gerontology Laboratory, Department of Biochemistry (DST-FIST Sponsored), Bharathidasan University Tiruchirappalli, India
| | - Sabapathy Periyasamy
- Molecular Gerontology Laboratory, Department of Biochemistry (DST-FIST Sponsored), Bharathidasan University Tiruchirappalli, India
| | - Kesavan S Jayachandran
- Molecular Cardiology and Drug Discovery Laboratory, Department of Bioinformatics, Bharathidasan University Tiruchirappalli, India
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Laboratory, Department of Biochemistry (DST-FIST Sponsored), Bharathidasan University Tiruchirappalli, India
| |
Collapse
|
32
|
Woodling NS, Andreasson KI. Untangling the Web: Toxic and Protective Effects of Neuroinflammation and PGE2 Signaling in Alzheimer's Disease. ACS Chem Neurosci 2016; 7:454-63. [PMID: 26979823 DOI: 10.1021/acschemneuro.6b00016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The neuroinflammatory response has received increasing attention as a key factor in the pathogenesis of Alzheimer's disease (AD). Microglia, the innate immune cells and resident phagocytes of the brain, respond to accumulating Aβ peptides by generating a nonresolving inflammatory response. While this response can clear Aβ peptides from the nervous system in some settings, its failure to do so in AD accelerates synaptic injury, neuronal loss, and cognitive decline. The complex molecular components of this response are beginning to be unraveled, with identification of both damaging and protective roles for individual components of the neuroinflammatory response. Even within one molecular pathway, contrasting effects are often present. As one example, recent studies of the inflammatory cyclooxygenase-prostaglandin pathway have revealed both beneficial and detrimental effects dependent on the disease context, cell type, and downstream signaling pathway. Nonsteroidal anti-inflammatory drugs (NSAIDs), which inhibit cyclooxygenases, are associated with reduced AD risk when taken by cognitively normal populations, but additional clinical and mouse model studies have added complexities and caveats to this finding. Downstream of cyclooxygenase activity, prostaglandin E2 signaling exerts both damaging pro-inflammatory and protective anti-inflammatory effects through actions of specific E-prostanoid G-protein coupled receptors on specific cell types. These complexities underscore the need for careful study of individual components of the neuroinflammatory response to better understand their contribution to AD pathogenesis and progression.
Collapse
Affiliation(s)
- Nathaniel S. Woodling
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| | - Katrin I. Andreasson
- Department of Neurology and
Neurological Sciences, Stanford University School of Medicine, 1201
Welch Road, Stanford, California 94305, United States
| |
Collapse
|
33
|
Hu S, Maiti P, Ma Q, Zuo X, Jones MR, Cole GM, Frautschy SA. Clinical development of curcumin in neurodegenerative disease. Expert Rev Neurother 2016; 15:629-37. [PMID: 26035622 DOI: 10.1586/14737175.2015.1044981] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Curcumin, a polyphenolic antioxidant derived from the turmeric root has undergone extensive preclinical development, showing remarkable efficacy in wound repair, cancer and inflammatory disorders. This review addresses the rationale for its use in neurodegenerative disease, particularly Alzheimer's disease. Curcumin is a pleiotropic molecule, which not only directly binds to and limits aggregation of the β-sheet conformations of amyloid characteristic of many neurodegenerative diseases but also restores homeostasis of the inflammatory system, boosts the heat shock system to enhance clearance of toxic aggregates, scavenges free radicals, chelates iron and induces anti-oxidant response elements. Although curcumin corrects dysregulation of multiple pathways, it may exert many effects via a few molecular targets. Pharmaceutical development of natural compounds like curcumin and synthetic derivatives have strong scientific rationale, but will require overcoming various hurdles including; high cost of trials, concern about profitability and misconceptions about drug specificity, stability, and bioavailability.
Collapse
Affiliation(s)
- Shuxin Hu
- Greater Los Angeles Healthcare System, Veteran's Administration, Geriatric Research Education and Clinical Center, Los Angeles, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Lim S, Choi JG, Moon M, Kim HG, Lee W, Bak HR, Sung H, Park CH, Kim SY, Oh MS. An Optimized Combination of Ginger and Peony Root Effectively Inhibits Amyloid-β Accumulation and Amyloid-β-Mediated Pathology in AβPP/PS1 Double-Transgenic Mice. J Alzheimers Dis 2016; 50:189-200. [PMID: 26639976 DOI: 10.3233/jad-150839] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The progressive aggregation of amyloid-β protein (Aβ) into senile plaques is a major pathological factor of Alzheimer's disease (AD) and is believed to result in memory impairment. We aimed to investigate the effect of an optimized combination of ginger and peony root (OCGP), a standardized herbal mixture of ginger and peony root, on Aβ accumulation and memory impairment in amyloid-β protein precursor (AβPP)/presenilin 1 (PS1) double-transgenic mice. In an in vitro thioflavin T fluorescence assay, 100 μg/ml OCGP inhibited Aβ accumulation to the same extent as did 10 μM curcumin. Furthermore, AβPP/PS1 double-transgenic mice treated with OCGP (50 or 100 mg/kg/day given orally for 14 weeks) exhibited reduced Aβ plaque accumulation in the hippocampus and lower levels of glial fibrillary acid protein and cyclooxygease-2 expression compared with vehicle-treated controls. These results suggest that OCGP may prevent memory impairment in AD by inhibiting Aβ accumulation and inflammation in the brain.
Collapse
Affiliation(s)
- Soonmin Lim
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Jin Gyu Choi
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Hyo Geun Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Wonil Lee
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Hyoung-Rok Bak
- CJ Healthcare R&D center, Majang-myeon, Icheon, Gyeonggi, Republic of Korea
| | - Hachang Sung
- CJ Healthcare R&D center, Majang-myeon, Icheon, Gyeonggi, Republic of Korea
| | - Chi Hye Park
- CJ Healthcare R&D center, Majang-myeon, Icheon, Gyeonggi, Republic of Korea
| | - Sun Yeou Kim
- Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-gu, Incheon, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Science, Graduate School, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
- Department of Oriental Pharmaceutical Science, College of Pharmacy and Kyung Hee East-West Pharmaceutical Research Institute, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| |
Collapse
|
35
|
Peña-Altamira E, Prati F, Massenzio F, Virgili M, Contestabile A, Bolognesi ML, Monti B. Changing paradigm to target microglia in neurodegenerative diseases: from anti-inflammatory strategy to active immunomodulation. Expert Opin Ther Targets 2015; 20:627-40. [PMID: 26568363 DOI: 10.1517/14728222.2016.1121237] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The importance of microglia in most neurodegenerative pathologies, from Parkinson's disease to amyotrophic lateral sclerosis and Alzheimer's disease, is increasingly recognized. Until few years ago, microglial activation in pathological conditions was considered dangerous to neurons due to its causing inflammation. Today we know that these glial cells also play a crucial physiological and neuroprotective role, which is altered in neurodegenerative conditions. AREAS COVERED The neuroinflammatory hypothesis for neurodegenerative diseases has led to the trial of anti-inflammatory agents as therapeutics with largely disappointing results. New information about the physiopathological role of microglia has highlighted the importance of immunomodulation as a potential new therapeutic approach. This review summarizes knowledge on microglia as a potential therapeutic target in the most common neurodegenerative diseases, with focus on compounds directed toward the modulation of microglial immune response through specific molecular pathways. EXPERT OPINION Here we support the innovative concept of targeting microglial cells by modulating their activity, rather than simply trying to counteract their inflammatory neurotoxicity, as a potential therapeutic approach for neurodegenerative diseases. The advantage of this therapeutic approach could be to reduce neuroinflammation and toxicity, while at the same time strengthening intrinsic neuroprotective properties of microglia and promoting neuroregeneration.
Collapse
Affiliation(s)
| | - Federica Prati
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Francesca Massenzio
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Marco Virgili
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Antonio Contestabile
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Maria Laura Bolognesi
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| | - Barbara Monti
- a Department of Pharmacy and Biotechnology , University of Bologna , Bologna , Italy
| |
Collapse
|
36
|
Syed H, Ikram MF, Yaqinuddin A, Ahmed T. Cyclooxygenase I and II inhibitors distinctly enhance hippocampal- and cortex-dependent cognitive functions in mice. Mol Med Rep 2015; 12:7649-56. [PMID: 26398269 DOI: 10.3892/mmr.2015.4351] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 08/17/2015] [Indexed: 11/06/2022] Open
Abstract
Cyclooxygenase (COX) enzymes are expressed in the brain; however, their role in hippocampus-dependent and cortex-dependent cognitive functions remains to be fully elucidated. The aim of the present study was to comparatively investigate the effects of piroxicam, a selective COX-I inhibitor, and celecoxib, a selective COX‑II inhibitor, on cognitive functions in an AlCl3‑induced neurotoxicity mouse model to understand the specific role of each COX enzyme in the hippocampus and cortex. The AlCl3 (250 mg/kg) was administered to the mice in drinking water and the drugs were administered in feed for 30 days. Assessments of memory, including a Morris water maze, social behavior and nesting behavior were performed in control and treated mice. The RNA expression of the COX enzymes were analyzed using reverse transcription‑quantitative polymerase chain reaction analysis. An ex‑vivo 2,2‑Diphenyl‑1‑picrylhydrazyl assay was performed in the hippocampus and cortex. Following 30 days of treatment with thedrugs, the mice in the celecoxib‑ and piroxicam‑treated groups exhibited enhanced learning (6.84 ± 0.76 and 9.20 ± 1.08, respectively), compared with the AlCl3‑induced neurotoxicity group (21.14 ± 0.76) on the fifth day of the Morris water maze test. Celecoxib treatment improved social affiliation in the AlCl3‑induced neurotoxicity group, the results of which were superior to piroxicam. Piroxicam led to better improvement in nesting score in the AlCl3‑induced neurotoxicity group. Both drugs decreased the expression levels of COX‑I and COX‑II in the hippocampus and cortex, and rescued oxidative stress levels. These findings suggested that each drug distinctly affected cognitive functions, highlighting the distinctive roles of COX-I and COX-II in learning and memory.
Collapse
Affiliation(s)
- Huma Syed
- Neurobiology Laboratory, Atta‑ur‑Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | | | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Touqeer Ahmed
- Neurobiology Laboratory, Atta‑ur‑Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| |
Collapse
|
37
|
Shadfar S, Hwang CJ, Lim MS, Choi DY, Hong JT. Involvement of inflammation in Alzheimer’s disease pathogenesis and therapeutic potential of anti-inflammatory agents. Arch Pharm Res 2015; 38:2106-19. [DOI: 10.1007/s12272-015-0648-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/08/2015] [Indexed: 02/06/2023]
|
38
|
Johansson JU, Woodling NS, Shi J, Andreasson KI. Inflammatory Cyclooxygenase Activity and PGE 2 Signaling in Models of Alzheimer's Disease. ACTA ACUST UNITED AC 2015; 11:125-131. [PMID: 28413375 PMCID: PMC5384338 DOI: 10.2174/1573395511666150707181414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/03/2015] [Accepted: 04/19/2015] [Indexed: 11/28/2022]
Abstract
The inflammatory response is a fundamental driving force in the pathogenesis of Alzheimer’s disease (AD). In the setting of accumulating immunogenic Aß peptide assemblies, microglia, the innate immune cells of the brain, generate a non-resolving immune response and fail to adequately clear accumulating Aß peptides, accelerating neuronal and synaptic injury. Pathological, biomarker, and imaging studies point to a prominent role of the innate immune response in AD development, and the molecular components of this response are beginning to be unraveled. The inflammatory cyclooxygenase-PGE2 pathway is implicated in pre-clinical development of AD, both in epidemiology of normal aging populations and in transgenic mouse models of Familial AD. The cyclooxygenase-PGE2 pathway modulates the inflammatory response to accumulating Aß peptides through actions of specific E-prostanoid G-protein coupled receptors.
Collapse
Affiliation(s)
- Jenny U Johansson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: SRI International, Menlo Park, CA, USA
| | - Nathaniel S Woodling
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: Institute of Healthy Ageing, University College London, London, UK
| | - Ju Shi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.,Present address: True North Therapeutics, South San Francisco, CA, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
39
|
Harris CJ, Voss K, Murchison C, Ralle M, Frahler K, Carter R, Rhoads A, Lind B, Robinson E, Quinn JF. Oral zinc reduces amyloid burden in Tg2576 mice. J Alzheimers Dis 2015; 41:179-92. [PMID: 24595193 DOI: 10.3233/jad-131703] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aggregation of amyloid-β in Alzheimer's disease can be affected by free transition metals such as copper and zinc in the brain. Addition of copper and zinc with amyloid acts to increase aggregation and copper additionally promotes the formation of reactive oxygen species. We propose that reduction of brain copper by blocking uptake of copper from the diet is a viable strategy to regulate the formation of insoluble amyloid-β in the brain of Tg2576 mice. Mice were treated with regimens of zinc acetate, which acts with metallothionein to block copper uptake in the gut, at various times along their lifespan to model prevention and treatment paradigms. We found that the mice tolerated zinc acetate well over the six month course of study. While we did not observe significant changes in cognition and behavior, there was a reduction in insoluble amyloid-β in the brain. This observation coincided with a reduction in brain copper and interestingly no change in brain zinc. Our findings show that blocking copper uptake from the diet can redistribute copper from the brain and reduce amyloid-β aggregation.
Collapse
Affiliation(s)
- Christopher J Harris
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Kellen Voss
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Charles Murchison
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, OR, USA
| | - Kate Frahler
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Raina Carter
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Allison Rhoads
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Betty Lind
- Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| | - Emily Robinson
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), Portland Veterans Affairs Medical Center, Portland, OR, USA Department of Neurology, Oregon Health and Sciences University, Portland, OR, USA
| |
Collapse
|
40
|
Abstract
INTRODUCTION Activated microglia are associated with the progression of Alzheimer's disease (AD), as well as many other neurodegenerative diseases of aging. Microglia are therefore key targets for therapeutic intervention. AREAS COVERED β-amyloid (Aβ) deposits activate the complement system, which, in turn, stimulates microglia to release neurotoxic materials. Research has focused primarily on anti-inflammatory agents to temper this toxic effect. More recently there has been a focus on converting microglia from this M1 state to an M2 state in which the toxic effects are reduced and their phagocytic activity toward Aβ enhanced. Studies in transgenic mice have suggested a number of possible anti-inflammatory approaches but they may not always be a good model. An example is vaccination with antibodies to Aβ, which is effective in mouse models, but has repeatedly failed in clinical trials. Biomarker studies indicate that AD commences many years prior to clinical onset. EXPERT OPINION A hopeful approach to a disease-modifying treatment of AD is to administer agents that inhibit the inflammatory stimulation of microglia or successfully convert them to an M2 state. However, any such treatment must be started early in the disease.
Collapse
Affiliation(s)
- Patrick L McGeer
- University of British Columbia, Kinsmen Laboratory of Neurological Research , 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3 , Canada
| | | |
Collapse
|
41
|
Ma QL, Yang F, Frautschy SA, Cole GM. PAK in Alzheimer disease, Huntington disease and X-linked mental retardation. CELLULAR LOGISTICS 2014; 2:117-125. [PMID: 23162743 PMCID: PMC3490962 DOI: 10.4161/cl.21602] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Developmental cognitive deficits including X-linked mental retardation (XLMR) can be caused by mutations in P21-activated kinase 3 (PAK3) that disrupt actin dynamics in dendritic spines. Neurodegenerative diseases such as Alzheimer disease (AD), where both PAK1 and PAK3 are dysregulated, may share final common pathways with XLMR. Independent of familial mutation, cognitive deficits emerging with aging, notably AD, begin after decades of normal function. This prolonged prodromal period involves the buildup of amyloid-β (Aβ) extracellular plaques and intraneuronal neurofibrillary tangles (NFT). Subsequently region dependent deficits in synapses, dendritic spines and cognition coincide with dysregulation in PAK1 and PAK. Specifically proximal to decline, cytoplasmic levels of actin-regulating Rho GTPase and PAK1 kinase are decreased in moderate to severe AD, while aberrant activation and translocation of PAK1 appears around the onset of cognitive deficits. Downstream to PAK1, LIM kinase inactivates cofilin, contributing to cofilin pathology, while the activation of Rho-dependent kinase ROCK increases Aβ production. Aβ activation of fyn disrupts neuronal PAK1 and ROCK-mediated signaling, resulting in synaptic deficits. Reductions in PAK1 by the anti-amyloid compound curcumin suppress synaptotoxicity. Similarly other neurological disorders, including Huntington disease (HD) show dysregulation of PAKs. PAK1 modulates mutant huntingtin toxicity by enhancing huntingtin aggregation, and inhibition of PAK activity protects HD as well as fragile X syndrome (FXS) symptoms. Since PAK plays critical roles in learning and memory and is disrupted in many cognitive disorders, targeting PAK signaling in AD, HD and XLMR may be a novel common therapeutic target for AD, HD and XLMR.
Collapse
Affiliation(s)
- Qiu-Lan Ma
- Department of Neurology; University of California Los Angeles; Los Angeles, CA USA ; Geriatric Research and Clinical Center; Greater Los Angeles Veterans Affairs Healthcare System; West Los Angeles Medical Center; Los Angeles, CA USA
| | | | | | | |
Collapse
|
42
|
Choi JK, Carreras I, Aytan N, Jenkins-Sahlin E, Dedeoglu A, Jenkins BG. The effects of aging, housing and ibuprofen treatment on brain neurochemistry in a triple transgene Alzheimer's disease mouse model using magnetic resonance spectroscopy and imaging. Brain Res 2014; 1590:85-96. [PMID: 25301691 DOI: 10.1016/j.brainres.2014.09.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 09/09/2014] [Accepted: 09/27/2014] [Indexed: 01/18/2023]
Abstract
We investigated a triple transgene Alzheimer's disease (AD) mouse model that recapitulates many of the neurochemical, anatomic, pathologic and behavioral defects seen in human AD. We studied the mice as a function of age and brain region and investigated potential therapy with the non-steroidal anti-inflammatory drug ibuprofen. Magnetic resonance spectroscopy (MRS) showed alterations characteristic of AD (i.e. increased myo-inositol and decreased N-acetylaspartate (NAA)). Mice at 6 months of age showed an increase in myo-inositol in the hippocampus at a time when the Aβ is intracellular, but not in amygdala or cortex. Myo-inositol increased as a function of age in the amygdala, cortex and striatum while NAA decreased only in the hippocampus and cortex at 17-23 months of age. Ibuprofen protected the increase of myo-inositol at six months of age in the hippocampus, but had no effect at 17-23 months of age (a time when Aβ is extracellular). In vivo MRI and MRS showed that at 17-23 months of age there was a significant protective effect of ibuprofen on hippocampal volume and NAA loss. Together, these data show the following: the increase in myo-inositol occurs before the decrease in NAA in hippocampus but not cortex; the hippocampus shows earlier changes than does the amygdale or cortex consistent with earlier deposition of Aβ40-42 in the hippocampus and ibuprofen protects against multiple components of the AD pathology. These data also show a profound effect of housing on this particular mouse model.
Collapse
Affiliation(s)
- Ji-Kyung Choi
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Isabel Carreras
- Department of Veterans Affairs, VA Boston Healthcare System, Boston, MA 02130, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Nur Aytan
- Department of Veterans Affairs, VA Boston Healthcare System, Boston, MA 02130, USA; Neurology and Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Eric Jenkins-Sahlin
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Alpaslan Dedeoglu
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Veterans Affairs, VA Boston Healthcare System, Boston, MA 02130, USA; Neurology and Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Bruce G Jenkins
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
43
|
Wang P, Guan PP, Wang T, Yu X, Guo JJ, Wang ZY. Aggravation of Alzheimer's disease due to the COX-2-mediated reciprocal regulation of IL-1β and Aβ between glial and neuron cells. Aging Cell 2014; 13:605-615. [PMID: 24621265 PMCID: PMC4326948 DOI: 10.1111/acel.12209] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2014] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and displays the characteristics of chronic neurodegenerative disorders; amyloid plaques (AP) that contain amyloid β-protein (Aβ) accumulate in AD, which is also characterized by tau phosphorylation. Epidemiological evidence has demonstrated that long-term treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) markedly reduces the risk of AD by inhibiting the expression of cyclooxygenase 2 (COX-2). Although the levels of COX-2 and its metabolic product prostaglandin (PG)E2 are elevated in the brain of AD patients, the mechanisms for the development of AD remain unknown. Using human- or mouse-derived glioblastoma and neuroblastoma cell lines as model systems, we delineated the signaling pathways by which COX-2 mediates the reciprocal regulation of interleukin-1β (IL-1β) and Aβ between glial and neuron cells. In glioblastoma cells, COX-2 regulates the synthesis of IL-1β in a PGE2 -dependent manner. Moreover, COX-2-derived PGE2 signals the activation of the PI3-K/AKT and PKA/CREB pathways via cyclic AMP; these pathways transactivate the NF-κB p65 subunit via phosphorylation at Ser 536 and Ser 276, leading to IL-1β synthesis. The secretion of IL-1β from glioblastoma cells in turn stimulates the expression of COX-2 in human or mouse neuroblastoma cells. Similar regulatory mechanisms were found for the COX-2 regulation of BACE-1 expression in neuroblastoma cells. More importantly, Aβ deposition mediated the inflammatory response of glial cells via inducing the expression of COX-2 in glioblastoma cells. These findings not only provide new insights into the mechanisms of COX-2-induced AD but also initially define the therapeutic targets of AD.
Collapse
Affiliation(s)
| | | | - Tao Wang
- College of Life and Health Sciences, Northeastern UniversityShenyang, 110819, China
| | - Xin Yu
- College of Life and Health Sciences, Northeastern UniversityShenyang, 110819, China
| | - Jian-Jun Guo
- College of Life and Health Sciences, Northeastern UniversityShenyang, 110819, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern UniversityShenyang, 110819, China
| |
Collapse
|
44
|
Takeda S, Sato N, Morishita R. Systemic inflammation, blood-brain barrier vulnerability and cognitive/non-cognitive symptoms in Alzheimer disease: relevance to pathogenesis and therapy. Front Aging Neurosci 2014; 6:171. [PMID: 25120476 PMCID: PMC4114193 DOI: 10.3389/fnagi.2014.00171] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/01/2014] [Indexed: 11/25/2022] Open
Abstract
The incidence of dementia is increasing at an alarming rate, and has become a major public health concern. Alzheimer disease (AD) is the most common form of dementia and is characterized by progressive cognitive impairment. In addition to classical neuropathological features such as amyloid plaques and neurofibrillary tangles (NFT), accumulation of activated immune cells has been documented in the AD brain, suggesting a contribution of neuroinflammation in the pathogenesis of AD. Besides cognitive deterioration, non-cognitive symptoms, such as agitation, aggression, depression and psychosis, are often observed in demented patients, including those with AD, and these neuropsychological symptoms place a heavy burden on caregivers. These symptoms often exhibit sudden onset and tend to fluctuate over time, and in many cases, they are triggered by an infection in peripheral organs, suggesting that inflammation plays an important role in the pathogenesis of these non-cognitive symptoms. However, there is no mechanistic explanation for the relationship between inflammation and neuropsychiatric symptoms. Observations from experimental mouse models indicate that alteration of brain blood vessels, especially blood-brain barrier (BBB) dysfunction, may contribute to the relationship. The current review summarizes the results from recent studies on the relationship between inflammation and AD, while focusing on cerebrovascular alterations, which might provide an insight into the pathogenesis of cognitive/non-cognitive symptoms in AD patients and suggest a basis for the development of new therapeutic treatments for these conditions.
Collapse
Affiliation(s)
- Shuko Takeda
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Yamadaoka Suita, Osaka, Japan ; Department of Geriatric Medicine, Graduate School of Medicine, Osaka University, Yamada-oka Suita, Osaka, Japan
| | - Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Yamadaoka Suita, Osaka, Japan ; Department of Geriatric Medicine, Graduate School of Medicine, Osaka University, Yamada-oka Suita, Osaka, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka University, Yamadaoka Suita, Osaka, Japan
| |
Collapse
|
45
|
Tracking neuroinflammation in Alzheimer's disease: the role of positron emission tomography imaging. J Neuroinflammation 2014; 11:120. [PMID: 25005532 PMCID: PMC4099095 DOI: 10.1186/1742-2094-11-120] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/20/2014] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) has been reconceptualized as a dynamic pathophysiological process, where the accumulation of amyloid-beta (Aβ) is thought to trigger a cascade of neurodegenerative events resulting in cognitive impairment and, eventually, dementia. In addition to Aβ pathology, various lines of research have implicated neuroinflammation as an important participant in AD pathophysiology. Currently, neuroinflammation can be measured in vivo using positron emission tomography (PET) with ligands targeting diverse biological processes such as microglial activation, reactive astrocytes and phospholipase A2 activity. In terms of therapeutic strategies, despite a strong rationale and epidemiological studies suggesting that the use of non-steroidal anti-inflammatory drugs (NSAIDs) may reduce the prevalence of AD, clinical trials conducted to date have proven inconclusive. In this respect, it has been hypothesized that NSAIDs may only prove protective if administered early on in the disease course, prior to the accumulation of significant AD pathology. In order to test various hypotheses pertaining to the exact role of neuroinflammation in AD, studies in asymptomatic carriers of mutations deterministic for early-onset familial AD may prove of use. In this respect, PET ligands for neuroinflammation may act as surrogate markers of disease progression, allowing for the development of more integrative models of AD, as well as for the measuring of target engagement in the context of clinical trials using NSAIDs. In this review, we address the biological basis of neuroinflammatory changes in AD, underscore therapeutic strategies using anti-inflammatory compounds, and shed light on the possibility of tracking neuroinflammation in vivo using PET imaging ligands.
Collapse
|
46
|
Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer's disease. Pharmacol Ther 2014; 142:244-57. [DOI: 10.1016/j.pharmthera.2013.12.009] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
|
47
|
Dong Z, Yan L, Huang G, Zhang L, Mei B, Meng B. Ibuprofen partially attenuates neurodegenerative symptoms in presenilin conditional double-knockout mice. Neuroscience 2014; 270:58-68. [PMID: 24699228 DOI: 10.1016/j.neuroscience.2014.03.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 03/06/2014] [Accepted: 03/23/2014] [Indexed: 01/22/2023]
Abstract
Ibuprofen is a widely used nonsteroidal anti-inflammatory drug that reportedly reduces the risk of Alzheimer's disease (AD) development. The anti-inflammatory effect of ibuprofen occurred via inhibition of cyclooxygenases and anti-amyloidogenesis through modulation of γ-secretase. Presenilin 1 and 2 conditional double-knockout (cDKO) mice exhibited age-dependent memory impairment and forebrain degeneration without elevation of amyloid β deposition. Therefore, cDKO mice can be an ideal animal model on which to independently test the effects of ibuprofen anti-inflammatory properties on the prevention of AD. Three- and six-month-old cDKO mice were fed diet containing 375 ppm ibuprofen for six months. After multiple, well-validated behavioral tests, treatment with ibuprofen improved cognition-related behavioral performance, and drug efficacy was correlated with the timing of administration. Ibuprofen was more effective on six-month-old than on three-month-old cDKO mice. Biochemical analysis demonstrated that the effects of ibuprofen on glial fibrillary acidic protein and CD68 expression levels were uneven in different brain regions of cDKO mice and that age also influenced such effects. Tau hyperphosphorylation and the cleavage of caspase-3 decreased after ibuprofen treatment, and this effect was more significant in the older than the younger group of mice, which was consistent with the results of behavioral tests.
Collapse
Affiliation(s)
- Z Dong
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - L Yan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - G Huang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - L Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - B Mei
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | - B Meng
- Key Laboratory of Brain Functional Genomics, Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
48
|
Birch AM, Katsouri L, Sastre M. Modulation of inflammation in transgenic models of Alzheimer's disease. J Neuroinflammation 2014; 11:25. [PMID: 24490742 PMCID: PMC3922595 DOI: 10.1186/1742-2094-11-25] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 01/21/2014] [Indexed: 12/23/2022] Open
Abstract
Over the past decade the process of inflammation has been a focus of increasing interest in the Alzheimer’s disease (AD) field, not only for its potential role in neuronal degeneration but also as a promising therapeutic target. However, recent research in this field has provided divergent outcomes, largely due to the use of different models and different stages of the disease when the investigations have been carried out. It is now accepted that microglia, and possibly astrocytes, change their activation phenotype during ageing and the stage of the disease, and therefore these are important factors to have in mind to define the function of different inflammatory components as well as potential therapies. Modulating inflammation using animal models of AD has offered the possibility to investigate inflammatory components individually and manipulate inflammatory genes in amyloid precursor protein and tau transgenics independently. This has also offered some hints on the mechanisms by which these factors may affect AD pathology. In this review we examine the different transgenic approaches and treatments that have been reported to modulate inflammation using animal models of AD. These studies have provided evidence that enhancing inflammation is linked with increases in amyloid-beta (Aβ) generation, Aβ aggregation and tau phosphorylation. However, the alterations on tau phosphorylation can be independent of changes in Aβ levels by these inflammatory mediators.
Collapse
Affiliation(s)
| | | | - Magdalena Sastre
- Division of Brain Sciences, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
49
|
Cudaback E, Jorstad NL, Yang Y, Montine TJ, Keene CD. Therapeutic implications of the prostaglandin pathway in Alzheimer's disease. Biochem Pharmacol 2014; 88:565-72. [PMID: 24434190 DOI: 10.1016/j.bcp.2013.12.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/18/2013] [Accepted: 12/18/2013] [Indexed: 11/19/2022]
Abstract
An important pathologic hallmark of Alzheimer's disease (AD) is neuroinflammation, a process characterized in AD by disproportionate activation of cells (microglia and astrocytes, primarily) of the non-specific innate immune system within the CNS. While inflammation itself is not intrinsically detrimental, a delicate balance of pro- and anti-inflammatory signals must be maintained to ensure that long-term exaggerated responses do not damage the brain over time. Non-steroidal anti-inflammatory drugs (NSAIDs) represent a broad class of powerful therapeutics that temper inflammation by inhibiting cyclooxygenase-mediated signaling pathways including prostaglandins, which are the principal mediators of CNS neuroinflammation. While historically used to treat discrete or systemic inflammatory conditions, epidemiologic evidence suggests that protracted NSAID use may delay AD onset, as well as decrease disease severity and rate of progression. Unfortunately, clinical trials with NSAIDs have thus far yielded disappointing results, including premature discontinuation of a large-scale prevention trial due to unexpected cardiovascular side effects. Here we review the literature and make the argument that more targeted exploitation of downstream prostaglandin signaling pathways may offer significant therapeutic benefits for AD while minimizing adverse side effects. Directed strategies such as these may ultimately help to delay the deleterious consequences of brain aging and might someday lead to new therapies for AD and other chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Eiron Cudaback
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Nikolas L Jorstad
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Yue Yang
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Thomas J Montine
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - C Dirk Keene
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA.
| |
Collapse
|
50
|
Attaby FA, Abdel-Fattah AM, Shaif LM, Elsayed MM. Synthesis, Reactions, Characterization and Biological Evaluation of 2,3′-Bipyridine Derivatives (III). J Heterocycl Chem 2013. [DOI: 10.1002/jhet.1521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Fawzy A. Attaby
- Chemistry Department, Faculty of Science; Cairo University; Giza 12613 Egypt
| | | | - Labeeb M. Shaif
- Chemistry Department, Faculty of Science; Cairo University; Giza 12613 Egypt
| | | |
Collapse
|