101
|
Biringer RG. The Role of Eicosanoids in Alzheimer's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16142560. [PMID: 31323750 PMCID: PMC6678666 DOI: 10.3390/ijerph16142560] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/09/2019] [Accepted: 07/13/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders known. Estimates from the Alzheimer's Association suggest that there are currently 5.8 million Americans living with the disease and that this will rise to 14 million by 2050. Research over the decades has revealed that AD pathology is complex and involves a number of cellular processes. In addition to the well-studied amyloid-β and tau pathology, oxidative damage to lipids and inflammation are also intimately involved. One aspect all these processes share is eicosanoid signaling. Eicosanoids are derived from polyunsaturated fatty acids by enzymatic or non-enzymatic means and serve as short-lived autocrine or paracrine agents. Some of these eicosanoids serve to exacerbate AD pathology while others serve to remediate AD pathology. A thorough understanding of eicosanoid signaling is paramount for understanding the underlying mechanisms and developing potential treatments for AD. In this review, eicosanoid metabolism is examined in terms of in vivo production, sites of production, receptor signaling, non-AD biological functions, and known participation in AD pathology.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd., Bradenton, FL 34211, USA.
| |
Collapse
|
102
|
Zhang X, Ye P, Wang D, Liu Y, Cao L, Wang Y, Xu Y, Zhu C. Involvement of RhoA/ROCK Signaling in Aβ-Induced Chemotaxis, Cytotoxicity and Inflammatory Response of Microglial BV2 Cells. Cell Mol Neurobiol 2019; 39:637-650. [PMID: 30852720 DOI: 10.1007/s10571-019-00668-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/28/2019] [Indexed: 12/14/2022]
Abstract
Reactive microglia clustering around amyloid plaques in brain is a histopathological feature of Alzheimer's disease (AD) and reflects the contribution of neuroinflammation in AD pathogenesis. β-Amyloid peptide (Aβ) has been shown to induce a range of microglial responses including chemotaxis, cytotoxicity and inflammation, but the underlying mechanism is poorly understood. Considering the fundamental role of RhoA/ROCK signaling in cell migration and its broad implication in AD and neuroinflammation, we hypothesized that RhoA/ROCK signaling might be involved in Aβ-induced microglial responses. From in vivo mouse models including APP/PS1 transgene and fibrillar Aβ stereotactic injection, we observed the elevated expression level of RhoA in reactive microglia. Through a series in vitro cell migration, cytotoxicity and biochemistry assays, we found that RhoA/ROCK signaling plays an essential role in Aβ-induced responses of microglial BV2 cells. Small molecular agents Fasudil and Y27632 showed prominent beneficial effects, which implies the therapeutic potential of RhoA/ROCK signaling inhibitors in AD treatment.
Collapse
Affiliation(s)
- Xiaoxu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Piao Ye
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Dandan Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Yunsheng Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Lan Cao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Yancong Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China
| | - Yuxia Xu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China.
| | - Cuiqing Zhu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai, 200032, China.
| |
Collapse
|
103
|
Yao K, Zu HB. Microglial polarization: novel therapeutic mechanism against Alzheimer's disease. Inflammopharmacology 2019; 28:95-110. [PMID: 31264132 DOI: 10.1007/s10787-019-00613-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease that results in progressive dementia, and exhibits high disability and fatality rates. Recent evidence has demonstrated that neuroinflammation is critical in the pathophysiological processes of AD, which is characterized by the activation of microglia and astrocytes. Under different stimuli, microglia are usually activated into two polarized states, termed the classical 'M1' phenotype and the alternative 'M2' phenotype. M1 microglia are considered to promote inflammatory injury in AD; in contrast, M2 microglia exert neuroprotective effects. Imbalanced microglial polarization, in the form of excessive activation of M1 microglia and dysfunction of M2 microglia, markedly promotes the development of AD. Furthermore, an increasing number of studies have shown that the transition of microglia from the M1 to M2 phenotype could potently alleviate pathological damage in AD. Hence, this article reviews the current knowledge regarding the role of microglial M1/M2 polarization in the pathophysiology of AD. In addition, we summarize several approaches that protect against AD by altering the polarization states of microglia. This review aims to contribute to a better understanding of the pathogenesis of AD and, moreover, to explore the potential of novel drugs for the treatment of AD in the future.
Collapse
Affiliation(s)
- Kai Yao
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China
| | - Heng-Bing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
104
|
Venkat P, Chopp M, Zacharek A, Cui C, Landschoot-Ward J, Qian Y, Chen Z, Chen J. Sildenafil treatment of vascular dementia in aged rats. Neurochem Int 2019; 127:103-112. [DOI: 10.1016/j.neuint.2018.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/24/2018] [Accepted: 12/24/2018] [Indexed: 01/08/2023]
|
105
|
Refolo V, Stefanova N. Neuroinflammation and Glial Phenotypic Changes in Alpha-Synucleinopathies. Front Cell Neurosci 2019; 13:263. [PMID: 31263402 PMCID: PMC6585624 DOI: 10.3389/fncel.2019.00263] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023] Open
Abstract
The role of neuroinflammation has been increasingly recognized in the field of neurodegenerative diseases. Many studies focusing on the glial cells involved in the inflammatory responses of the brain, namely microglia and astroglia, have over the years pointed out the dynamic and changing behavior of these cells, accompanied by different morphologies and activation forms. This is particularly evident in diseased conditions, where glia react to any shift from homeostasis, acquiring different phenotypes. Particularly for microglia, it has soon become clear that such phenotypes are multiple, as multiple are the functions related to them. Several approaches have over time revealed different facets of microglial phenotypic diversity, and advanced genetic analyses, in recent years, have added new insights into microglial heterogeneity, opening novel scenarios that researchers have just started to explore. Among neurodegenerative diseases, an important section is represented by alpha-synucleinopathies. Here alpha-synuclein accumulates abnormally in the brain and, depending on its pattern of distribution, leads to the development of different clinical conditions. Also for these proteinopathies, neuroinflammation and glial activation have been identified as constant and crucial factors during disease development. In the present review we will address the current literature about glial phenotypic changes with respect to alpha-synucleinopathies, as well as consider the pathophysiological and therapeutic implications of such a dynamic cellular behavior.
Collapse
Affiliation(s)
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
106
|
Hsueh SC, Lecca D, Greig NH, Wang JY, Selman W, Hoffer BJ, Miller JP, Chiang YH. (-)-Phenserine Ameliorates Contusion Volume, Neuroinflammation, and Behavioral Impairments Induced by Traumatic Brain Injury in Mice. Cell Transplant 2019; 28:1183-1196. [PMID: 31177840 PMCID: PMC6767878 DOI: 10.1177/0963689719854693] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI), a major cause of mortality and morbidity, affects 10 million people worldwide, with limited treatment options. We have previously shown that (-)-phenserine (Phen), an acetylcholinesterase inhibitor originally designed and tested in clinical phase III trials for Alzheimer's disease, can reduce neurodegeneration after TBI and reduce cognitive impairments induced by mild TBI. In this study, we used a mouse model of moderate to severe TBI by controlled cortical impact to assess the effects of Phen on post-trauma histochemical and behavioral changes. Animals were treated with Phen (2.5 mg/kg, IP, BID) for 5 days started on the day of injury and the effects were evaluated by behavioral and histological examinations at 1 and 2 weeks after injury. Phen significantly attenuated TBI-induced contusion volume, enlargement of the lateral ventricle, and behavioral impairments in motor asymmetry, sensorimotor functions, motor coordination, and balance functions. The morphology of microglia was shifted to an active from a resting form after TBI, and Phen dramatically reduced the ratio of activated to resting microglia, suggesting that Phen also mitigates neuroinflammation after TBI. While Phen has potent anti-acetylcholinesterase activity, its (+) isomer Posiphen shares many neuroprotective properties but is almost completely devoid of anti-acetylcholinesterase activity. We evaluated Posiphen at a similar dose to Phen and found similar mitigation in lateral ventricular size increase, motor asymmetry, motor coordination, and balance function, suggesting the improvement of these histological and behavioral tests by Phen treatment occur via pathways other than anti-acetylcholinesterase inhibition. However, the reduction of lesion size and improvement of sensorimotor function by Posiphen were much smaller than with equivalent doses of Phen. Taken together, these results show that post-injury treatment with Phen over 5 days significantly ameliorates severity of TBI. These data suggest a potential development of this compound for clinical use in TBI therapy.
Collapse
Affiliation(s)
- Shih-Chang Hsueh
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniela Lecca
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jia-Yi Wang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
| | - Warren Selman
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Barry J Hoffer
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jonathan P Miller
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yung-Hsiao Chiang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei.,Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei
| |
Collapse
|
107
|
Chandra S, Pahan K. Gemfibrozil, a Lipid-Lowering Drug, Lowers Amyloid Plaque Pathology and Enhances Memory in a Mouse Model of Alzheimer's Disease via Peroxisome Proliferator-Activated Receptor α. J Alzheimers Dis Rep 2019; 3:149-168. [PMID: 31259309 PMCID: PMC6597963 DOI: 10.3233/adr-190104] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Deposition of extracellular senile plaques containing amyloid-β is one of the major neuropathological characteristics of Alzheimer’s disease (AD). Therefore, targeting amyloid-β dyshomeostasis is an important therapeutic strategy for treatment of AD. In this study, we demonstrate that gemfibrozil, an FDA-approved drug for hyperlipidemia, can lower the amyloid plaque burden in the hippocampus and cortex of the 5XFAD model of AD. Additionally, gemfibrozil reduced microgliosis and astrogliosis associated with plaque in these mice. Administration of gemfibrozil also improved spatial learning and memory of the 5XFAD mice. Finally, we delineate that gemfibrozil requires the transcription factor peroxisome proliferator-activated receptor alpha (PPARα) to exhibit its amyloid lowering and memory enhancing effects in 5XFAD mice. These results highlight a new therapeutic property of gemfibrozil and suggest that this drug may be repurposed for treatment of AD.
Collapse
Affiliation(s)
- Sujyoti Chandra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Kalipada Pahan
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.,Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
108
|
Purvis GSD, Solito E, Thiemermann C. Annexin-A1: Therapeutic Potential in Microvascular Disease. Front Immunol 2019; 10:938. [PMID: 31114582 PMCID: PMC6502989 DOI: 10.3389/fimmu.2019.00938] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Annexin-A1 (ANXA1) was first discovered in the early 1980's as a protein, which mediates (some of the) anti-inflammatory effects of glucocorticoids. Subsequently, the role of ANXA1 in inflammation has been extensively studied. The biology of ANXA1 is complex and it has many different roles in both health and disease. Its effects as a potent endogenous anti-inflammatory mediator are well-described in both acute and chronic inflammation and its role in activating the pro-resolution phase receptor, FPR2, has been described and is now being exploited for therapeutic benefit. In the present mini review, we will endeavor to give an overview of ANXA1 biology in relation to inflammation and functions that mediate pro-resolution that are independent of glucocorticoid induction. We will focus on the role of ANXA1 in diseases with a large inflammatory component focusing on diabetes and microvascular disease. Finally, we will explore the possibility of exploiting ANXA1 as a novel therapeutic target in diabetes and the treatment of microvascular disease.
Collapse
Affiliation(s)
- Gareth S D Purvis
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Egle Solito
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Christoph Thiemermann
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
109
|
Chung HY, Kim DH, Lee EK, Chung KW, Chung S, Lee B, Seo AY, Chung JH, Jung YS, Im E, Lee J, Kim ND, Choi YJ, Im DS, Yu BP. Redefining Chronic Inflammation in Aging and Age-Related Diseases: Proposal of the Senoinflammation Concept. Aging Dis 2019; 10:367-382. [PMID: 31011483 PMCID: PMC6457053 DOI: 10.14336/ad.2018.0324] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/24/2018] [Indexed: 12/13/2022] Open
Abstract
Age-associated chronic inflammation is characterized by unresolved and uncontrolled inflammation with multivariable low-grade, chronic and systemic responses that exacerbate the aging process and age-related chronic diseases. Currently, there are two major hypotheses related to the involvement of chronic inflammation in the aging process: molecular inflammation of aging and inflammaging. However, neither of these hypotheses satisfactorily addresses age-related chronic inflammation, considering the recent advances that have been made in inflammation research. A more comprehensive view of age-related inflammation, that has a scope beyond the conventional view, is therefore required. In this review, we discuss newly emerging data on multi-phase inflammatory networks and proinflammatory pathways as they relate to aging. We describe the age-related upregulation of nuclear factor (NF)-κB signaling, cytokines/chemokines, endoplasmic reticulum (ER) stress, inflammasome, and lipid accumulation. The later sections of this review present our expanded view of age-related senescent inflammation, a process we term "senoinflammation", that we propose here as a novel concept. As described in the discussion, senoinflammation provides a schema highlighting the important and ever-increasing roles of proinflammatory senescence-associated secretome, inflammasome, ER stress, TLRs, and microRNAs, which support the senoinflammation concept. It is hoped that this new concept of senoinflammation opens wider and deeper avenues for basic inflammation research and provides new insights into the anti-inflammatory therapeutic strategies targeting the multiple proinflammatory pathways and mediators and mediators that underlie the pathophysiological aging process.
Collapse
Affiliation(s)
- Hae Young Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Dae Hyun Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Eun Kyeong Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
- Pathological and Analytical Center, Korea Institute of Toxicology, Daejeon 34114, Korea.
| | - Ki Wung Chung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Sangwoon Chung
- Department of Internal Medicine, Pulmonary, Allergy, Critical Care & Sleep Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Bonggi Lee
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea.
| | - Arnold Y. Seo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Jae Heun Chung
- Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Korea.
| | - Young Suk Jung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Eunok Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Jaewon Lee
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Nam Deuk Kim
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Yeon Ja Choi
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, Korea.
| | - Dong Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 609-735, Korea.
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, TX 78229, USA.
| |
Collapse
|
110
|
Warren-Gash C, Forbes HJ, Williamson E, Breuer J, Hayward AC, Mavrodaris A, Ridha BH, Rossor MN, Thomas SL, Smeeth L. Human herpesvirus infections and dementia or mild cognitive impairment: a systematic review and meta-analysis. Sci Rep 2019; 9:4743. [PMID: 30894595 PMCID: PMC6426940 DOI: 10.1038/s41598-019-41218-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Interest is growing in the role of infectious agents in the pathogenesis of dementia, but current evidence is limited. We conducted a systematic review and meta-analysis to investigate the effect of any of eight human herpesviruses on development of dementia or mild cognitive impairment (MCI). We searched the Cochrane Library, Embase, Global Health, Medline, PsycINFO, Scopus, Web of Science, clinical trials registers and grey literature sources from inception to December 2017 for observational studies with cohort, case control or self-controlled designs, or randomised controlled trials of interventions against herpesviruses. Pooled effect estimates and 95% confidence intervals (CIs) were generated through random effects meta-analyses across studies with the same design, outcome, and virus type, method and site of measurement. We included 57 studies across various geographic settings. Past infection with herpesviruses, measured by IgG seropositivity, was generally not associated with dementia risk. A single cohort study rated moderate quality showed an association between varicella zoster virus reactivation (ophthalmic zoster) and incident dementia (HR 2.97; 95%CI, 1.89 to 4.66). Recent infection with, or reactivation of, herpes simplex virus type 1 or type 1/2 unspecified, cytomegalovirus and human herpes virus-6 measured by serum IgM, high titre IgG or clinical disease may be associated with dementia or MCI, though results were inconsistent across studies and overall evidence rated very low quality. Longitudinal population studies with robust repeated virus measurements taken sufficiently proximal to dementia onset are needed to establish whether, when and among whom herpesviruses affect dementia risk.
Collapse
Affiliation(s)
- Charlotte Warren-Gash
- Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom.
| | - Harriet J Forbes
- Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| | - Elizabeth Williamson
- Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| | - Judith Breuer
- Division of Infection & Immunity, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Andrew C Hayward
- Institute of Epidemiology and Healthcare, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Angelique Mavrodaris
- Cambridge Institute of Public Health, University of Cambridge, Forvie site, Robinson Way, Cambridge, CB2 0SR, United Kingdom
| | - Basil H Ridha
- NIHR University College London Hospitals Biomedical Research Centre, Maple House, Tottenham Court Road, London, W1T 7DN, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, United Kingdom
| | - Martin N Rossor
- NIHR University College London Hospitals Biomedical Research Centre, Maple House, Tottenham Court Road, London, W1T 7DN, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, United Kingdom
| | - Sara L Thomas
- Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| | - Liam Smeeth
- Faculty of Epidemiology & Population Health, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| |
Collapse
|
111
|
Wang Q, Wang Y, Liu J, Sutphen CL, Cruchaga C, Blazey T, Gordon BA, Su Y, Chen C, Shimony JS, Ances BM, Cairns NJ, Fagan AM, Morris JC, Benzinger TLS. Quantification of white matter cellularity and damage in preclinical and early symptomatic Alzheimer's disease. Neuroimage Clin 2019; 22:101767. [PMID: 30901713 PMCID: PMC6428957 DOI: 10.1016/j.nicl.2019.101767] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/12/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023]
Abstract
Interest in understanding the roles of white matter (WM) inflammation and damage in the pathophysiology of Alzheimer disease (AD) has been growing significantly in recent years. However, in vivo magnetic resonance imaging (MRI) techniques for imaging inflammation are still lacking. An advanced diffusion-based MRI method, neuro-inflammation imaging (NII), has been developed to clinically image and quantify WM inflammation and damage in AD. Here, we employed NII measures in conjunction with cerebrospinal fluid (CSF) biomarker classification (for β-amyloid (Aβ) and neurodegeneration) to evaluate 200 participants in an ongoing study of memory and aging. Elevated NII-derived cellular diffusivity was observed in both preclinical and early symptomatic phases of AD, while disruption of WM integrity, as detected by decreased fractional anisotropy (FA) and increased radial diffusivity (RD), was only observed in the symptomatic phase of AD. This may suggest that WM inflammation occurs earlier than WM damage following abnormal Aβ accumulation in AD. The negative correlation between NII-derived cellular diffusivity and CSF Aβ42 level (a marker of amyloidosis) may indicate that WM inflammation is associated with increasing Aβ burden. NII-derived FA also negatively correlated with CSF t-tau level (a marker of neurodegeneration), suggesting that disruption of WM integrity is associated with increasing neurodegeneration. Our findings demonstrated the capability of NII to simultaneously image and quantify WM cellularity changes and damage in preclinical and early symptomatic AD. NII may serve as a clinically feasible imaging tool to study the individual and composite roles of WM inflammation and damage in AD.
Collapse
Affiliation(s)
- Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA
| | - Yong Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Engineering & Applied Science, St. Louis, MO 63015, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Jingxia Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Courtney L Sutphen
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carlos Cruchaga
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tyler Blazey
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA
| | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ 85006, USA
| | - Charlie Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beau M Ances
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne M Fagan
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park, Suite 101, St. Louis, MO 63108, USA; Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
112
|
In Vitro Priming and Hyper-Activation of Brain Microglia: an Assessment of Phenotypes. Mol Neurobiol 2019; 56:6409-6425. [DOI: 10.1007/s12035-019-1529-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022]
|
113
|
Rey C, Delpech JC, Madore C, Nadjar A, Greenhalgh AD, Amadieu C, Aubert A, Pallet V, Vaysse C, Layé S, Joffre C. Dietary n-3 long chain PUFA supplementation promotes a pro-resolving oxylipin profile in the brain. Brain Behav Immun 2019; 76:17-27. [PMID: 30086401 DOI: 10.1016/j.bbi.2018.07.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 11/26/2022] Open
Abstract
The brain is highly enriched in long chain polyunsaturated fatty acids (LC-PUFAs) that display immunomodulatory properties in the brain. At the periphery, the modulation of inflammation by LC-PUFAs occurs through lipid mediators called oxylipins which have anti-inflammatory and pro-resolving activities when derived from n-3 LC-PUFAs and pro-inflammatory activities when derived from n-6 LC-PUFAs. However, whether a diet rich in LC-PUFAs modulates oxylipins and neuroinflammation in the brain has been poorly investigated. In this study, the effect of a dietary n-3 LC-PUFA supplementation on oxylipin profile and neuroinflammation in the brain was analyzed. Mice were given diets deficient or supplemented in n-3 LC-PUFAs for a 2-month period starting at post-natal day 21, followed by a peripheral administration of lipopolysaccharide (LPS) at adulthood. We first showed that dietary n-3 LC-PUFA supplementation induced n-3 LC-PUFA enrichment in the hippocampus and subsequently an increase in n-3 PUFA-derived oxylipins and a decrease in n-6 PUFA-derived oxylipins. In response to LPS, n-3 LC-PUFA deficient mice presented a pro-inflammatory oxylipin profile whereas n-3 LC-PUFA supplemented mice displayed an anti-inflammatory oxylipin profile in the hippocampus. Accordingly, the expression of cyclooxygenase-2 and 5-lipoxygenase, the enzymes implicated in pro- and anti-inflammatory oxylipin synthesis, was induced by LPS in both diets. In addition, LPS-induced pro-inflammatory cytokine increase was reduced by dietary n-3 LC-PUFA supplementation. These results indicate that brain n-3 LC-PUFAs increase by dietary means and promote the synthesis of anti-inflammatory derived bioactive oxylipins. As neuroinflammation plays a key role in all brain injuries and many neurodegenerative disorders, the present data suggest that dietary habits may be an important regulator of brain cytokine production in these contexts.
Collapse
Affiliation(s)
- C Rey
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; ITERG, Institut des corps gras, 33600 Pessac, France
| | - J C Delpech
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - C Madore
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - A D Greenhalgh
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - C Amadieu
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - A Aubert
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - V Pallet
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - C Vaysse
- ITERG, Institut des corps gras, 33600 Pessac, France
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - C Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France.
| |
Collapse
|
114
|
Chen C, Wu S, Hong Z, Chen X, Shan X, Fischbach S, Xiao X. Chronic hyperglycemia regulates microglia polarization through ERK5. Aging (Albany NY) 2019; 11:697-706. [PMID: 30684443 PMCID: PMC6366978 DOI: 10.18632/aging.101770] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/07/2019] [Indexed: 05/01/2023]
Abstract
Diabetic patients are prone to developing Alzheimer's disease (AD), in which microglia play a critical role. However, the direct effect of high glucose (HG) on microglia and the role of extracellular-signal-regulated kinase 5 (ERK5) signaling in this interaction have not been examined before. Here, these questions were addressed in microglia cultured in HG versus normal glucose (NG) conditions. Initially, HG induced microglial differentiation into the M2a phenotype with concomitant ERK5 activation. However, longer exposure to HG further induced differentiation of microglia into the M2b-like phenotype, followed by the M1-like subtype, concomitant with a gradual loss of ERK5 activation. BIX021895, a specific inhibitor of ERK5 activation, prevented M2a- differentiation of microglia, but induced earlier M2b-like polarization followed by M1-like polarization. Transfection of microglia with a sustained activated form of MEK5 (MEK5DD) prolonged the duration of the M2a phenotype, and prevented later differentiation into the M2b/M1 subtype. Conditioned media from the M2a-polarized microglia reduced neuronal cell apoptosis in hypoxic condition, while media from M2b-like or M1-like microglia enhanced apoptosis. Together, our data suggest that chronic hyperglycemia may induce a gradual alteration of microglia polarization into an increasingly proinflammatory subtype, which could be suppressed by sustained activation of ERK5 signaling.
Collapse
Affiliation(s)
- Congde Chen
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou
Medical University, Wenzhou 325000, China
| | - Suichun Wu
- Reproductive Medicine Centre, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zipu Hong
- Department of Pediatric Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaoming Chen
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou
Medical University, Wenzhou 325000, China
- Department of Pediatric Endocrinology and Metabolism, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University , Wenzhou 325000, China
| | - Xiaoou Shan
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou
Medical University, Wenzhou 325000, China
- Department of Pediatric Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Shane Fischbach
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Xiangwei Xiao
- Department of Pediatric Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou
Medical University, Wenzhou 325000, China
- Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| |
Collapse
|
115
|
Nair S, Sobotka KS, Joshi P, Gressens P, Fleiss B, Thornton C, Mallard C, Hagberg H. Lipopolysaccharide-induced alteration of mitochondrial morphology induces a metabolic shift in microglia modulating the inflammatory response in vitro and in vivo. Glia 2019; 67:1047-1061. [PMID: 30637805 DOI: 10.1002/glia.23587] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that changes in the metabolic signature of microglia underlie their response to inflammation. We sought to increase our knowledge of how pro-inflammatory stimuli induce metabolic changes. Primary microglia exposed to lipopolysaccharide (LPS)-expressed excessive fission leading to more fragmented mitochondria than tubular mitochondria. LPS-mediated Toll-like receptor 4 (TLR4) activation also resulted in metabolic reprogramming from oxidative phosphorylation to glycolysis. Blockade of mitochondrial fission by Mdivi-1, a putative mitochondrial division inhibitor led to the reversal of the metabolic shift. Mdivi-1 treatment also normalized the changes caused by LPS exposure, namely an increase in mitochondrial reactive oxygen species production and mitochondrial membrane potential as well as accumulation of key metabolic intermediate of TCA cycle succinate. Moreover, Mdivi-1 treatment substantially reduced LPS induced cytokine and chemokine production. Finally, we showed that Mdivi-1 treatment attenuated expression of genes related to cytotoxic, repair, and immunomodulatory microglia phenotypes in an in vivo neuroinflammation paradigm. Collectively, our data show that the activation of microglia to a classically pro-inflammatory state is associated with a switch to glycolysis that is mediated by mitochondrial fission, a process which may be a pharmacological target for immunomodulation.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina S Sobotka
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pooja Joshi
- PROTECT, INSERM, Université Paris Diderot, Paris, France
| | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Bobbi Fleiss
- PROTECT, INSERM, Université Paris Diderot, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Claire Thornton
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Carina Mallard
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
116
|
Morris G, Berk M, Maes M, Puri BK. Could Alzheimer's Disease Originate in the Periphery and If So How So? Mol Neurobiol 2019; 56:406-434. [PMID: 29705945 PMCID: PMC6372984 DOI: 10.1007/s12035-018-1092-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
The classical amyloid cascade model for Alzheimer's disease (AD) has been challenged by several findings. Here, an alternative molecular neurobiological model is proposed. It is shown that the presence of the APOE ε4 allele, altered miRNA expression and epigenetic dysregulation in the promoter region and exon 1 of TREM2, as well as ANK1 hypermethylation and altered levels of histone post-translational methylation leading to increased transcription of TNFA, could variously explain increased levels of peripheral and central inflammation found in AD. In particular, as a result of increased activity of triggering receptor expressed on myeloid cells 2 (TREM-2), the presence of the apolipoprotein E4 (ApoE4) isoform, and changes in ANK1 expression, with subsequent changes in miR-486 leading to altered levels of protein kinase B (Akt), mechanistic (previously mammalian) target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), all of which play major roles in microglial activation, proliferation and survival, there is activation of microglia, leading to the subsequent (further) production of cytokines, chemokines, nitric oxide, prostaglandins, reactive oxygen species, inducible nitric oxide synthase and cyclooxygenase-2, and other mediators of inflammation and neurotoxicity. These changes are associated with the development of amyloid and tau pathology, mitochondrial dysfunction (including impaired activity of the electron transport chain, depleted basal mitochondrial potential and oxidative damage to key tricarboxylic acid enzymes), synaptic dysfunction, altered glycogen synthase kinase-3 (GSK-3) activity, mTOR activation, impairment of autophagy, compromised ubiquitin-proteasome system, iron dyshomeostasis, changes in APP translation, amyloid plaque formation, tau hyperphosphorylation and neurofibrillary tangle formation.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, Kenneth Myer Building, University of Melbourne, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
117
|
Bajwa E, Pointer CB, Klegeris A. The Role of Mitochondrial Damage-Associated Molecular Patterns in Chronic Neuroinflammation. Mediators Inflamm 2019; 2019:4050796. [PMID: 31065234 PMCID: PMC6466851 DOI: 10.1155/2019/4050796] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction has been established as a common feature of neurodegenerative disorders that contributes to disease pathology by causing impaired cellular energy production. Mitochondrial molecules released into the extracellular space following neuronal damage or death may also play a role in these diseases by acting as signaling molecules called damage-associated molecular patterns (DAMPs). Mitochondrial DAMPs have been shown to initiate proinflammatory immune responses from nonneuronal glial cells, including microglia and astrocytes; thereby, they have the potential to contribute to the chronic neuroinflammation present in these disorders accelerating the degeneration of neurons. In this review, we highlight the mitochondrial DAMPs cytochrome c (CytC), mitochondrial transcription factor A (TFAM), and cardiolipin and explore their potential role in the central nervous system disorders including Alzheimer's disease and Parkinson's disease, which are characterized by neurodegeneration and chronic neuroinflammation.
Collapse
Affiliation(s)
- Ekta Bajwa
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| | - Caitlin B. Pointer
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, Canada
| |
Collapse
|
118
|
Villa A, Della Torre S, Maggi A. Sexual differentiation of microglia. Front Neuroendocrinol 2019; 52:156-164. [PMID: 30481522 DOI: 10.1016/j.yfrne.2018.11.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/07/2018] [Accepted: 11/24/2018] [Indexed: 12/28/2022]
Abstract
Sex plays a role in the incidence and outcome of neurological illnesses, also influencing the response to treatments. Despite sexual differentiation of the brain has been extensively investigated, the study of sex differences in microglia, the brain's resident immune cells, has been largely neglected until recently. To fulfill this gap, our laboratory developed several tools, including cellular and animal models, which bolstered in-depth studies on sexual differentiation of microglia and its impact on brain physiology, as well as on the onset and progression of neurological disorders. Here, we summarize the current status of knowledge on the sex-dependent function of microglia, and report recent evidence linking these cells to the sexual bias in the susceptibility to neurological brain diseases.
Collapse
Affiliation(s)
- Alessandro Villa
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy
| | - Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases and Dept of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti, 9, Milan, Italy.
| |
Collapse
|
119
|
Martínez-Frailes C, Di Lauro C, Bianchi C, de Diego-García L, Sebastián-Serrano Á, Boscá L, Díaz-Hernández M. Amyloid Peptide Induced Neuroinflammation Increases the P2X7 Receptor Expression in Microglial Cells, Impacting on Its Functionality. Front Cell Neurosci 2019; 13:143. [PMID: 31031598 PMCID: PMC6474397 DOI: 10.3389/fncel.2019.00143] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/22/2019] [Indexed: 02/05/2023] Open
Abstract
Alzheimer disease is a neurodegenerative disease characterized by the presence of senile plaques composed of amyloid-β (Aβ) peptide, neurofibrillary tangles, neuronal loss and neuroinflammation. Previous works have revealed that extracellular ATP, through its selective receptor P2X7 (P2X7R), is essential to neuroinflammation and neurotoxicity induced by Aβ. P2X7R is upregulated on microglial cells around the senile plaques. This upregulation progressively rises with age and is parallel with an accumulation of senile plaques and also correlates with the synaptic toxicity detected both in animal models reproducing AD and human patients of AD. Furthermore, the late onset of the first AD-associated symptoms suggests that aging associated-changes may be relevant to the disease progression. Thus, microglia motility and its capacity to respond to exogenous ATP stimulus decrease with aging. To evaluate whether the P2X7R age related-changes on microglia cells may be relevant to the AD progression, we generated a new transgenic mouse model crossing an Aβ peptide mouse model, J20 mice and the P2X7R reporter mice P2X7REGFP. Our results indicate that neuroinflammation induced by Aβ peptide causes changes in the P2X7R distribution pattern, increasing it s expression in microglial cells at advanced and late stages, when microgliosis occurs, but not in the early stages, in the absence of microgliosis. In addition, we found that P2X7R activation promotes microglial cells migration to senile plaques but decreases their phagocytic capacity. Moreover, we found a significant reduction of P2X7R transcription on neuronal cells at the early and advanced stages, but not at the late stages. Since previous studies have reported that either pharmacological inhibition or selective downregulation of P2X7R significantly improve behavioral alterations and reduce the incidence and size of senile plaques in the early and advanced stages of AD, the results presented here provide new evidence, indicating that this therapeutic approach could be also efficient in the late stages of the disease.
Collapse
Affiliation(s)
- Carlos Martínez-Frailes
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Caterina Di Lauro
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Carolina Bianchi
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Laura de Diego-García
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
| | - Álvaro Sebastián-Serrano
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigaciones Biomedicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomedicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Díaz-Hernández
- Department of Biochemistry and Molecular Biology, Veterinary School, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, Spain
- *Correspondence: Miguel Díaz-Hernández,
| |
Collapse
|
120
|
Tournier BB, Tsartsalis S, Rigaud D, Fossey C, Cailly T, Fabis F, Pham T, Grégoire MC, Kövari E, Moulin-Sallanon M, Savioz A, Millet P. TSPO and amyloid deposits in sub-regions of the hippocampus in the 3xTgAD mouse model of Alzheimer’s disease. Neurobiol Dis 2019; 121:95-105. [DOI: 10.1016/j.nbd.2018.09.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/03/2018] [Accepted: 09/23/2018] [Indexed: 11/16/2022] Open
|
121
|
Radisavljevic N, Cirstea M, Brett Finlay B. Bottoms up: the role of gut microbiota in brain health. Environ Microbiol 2018; 21:3197-3211. [PMID: 30556271 DOI: 10.1111/1462-2920.14506] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 12/14/2022]
Abstract
The gut microbiota affects many aspects of human health, and research, especially over the past decade, is demonstrating that the brain is no exception. This review summarizes existing human observational studies of the microbiota in brain health and neurological conditions at all ages, as well as animal studies that are advancing the field beyond correlation and into causality. Potential mechanisms by which the brain and the gut microbiota are connected are explored, including inflammation, bacterially-produced metabolites and neurotransmitters and specific roles for individual microbes. Finally, important challenges and potential mitigation strategies are discussed, as well as ways in which some of these same challenges can be harnessed to advance our understanding of this complex, exciting and rapidly evolving field.
Collapse
Affiliation(s)
- Nina Radisavljevic
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Barton Brett Finlay
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
122
|
Luca A, Calandra C, Luca M. Molecular Bases of Alzheimer's Disease and Neurodegeneration: The Role of Neuroglia. Aging Dis 2018; 9:1134-1152. [PMID: 30574424 PMCID: PMC6284765 DOI: 10.14336/ad.2018.0201] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Neuroglia is an umbrella term indicating different cellular types that play a pivotal role in the brain, being involved in its development and functional homeostasis. Glial cells are becoming the focus of recent researches pertaining the pathogenesis of neurodegenerative disorders, Alzheimer's Disease (AD) in particular. In fact, activated microglia is the main determinant of neuroinflammation, contributing to neurodegeneration. In addition, the oxidative insult occurring during pathological brain aging can activate glial cells that, in turn, can favor the production of free radicals. Moreover, the recent Glycogen Synthase Kinase 3 (GSK-3) hypothesis of AD suggests that GSK3, involved in the regulation of glial cells functioning, could exert a role in amyloid deposition and tau hyper-phosphorylation. In this review, we briefly describe the main physiological functions of the glial cells and discuss the link between neuroglia and the most studied molecular bases of AD. In addition, we dedicate a section to the glial changes occurring in AD, with particular attention to their role in terms of neurodegeneration. In the light of the literature data, neuroglia could play a fundamental role in AD pathogenesis and progression. Further studies are needed to shed light on this topic.
Collapse
Affiliation(s)
- Antonina Luca
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Carmela Calandra
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| | - Maria Luca
- Department of General Surgery and Medical-Surgical Specialties, Dermatology Clinic, University Hospital Policlinico-Vittorio Emanuele, Catania, 95100 Sicily, Italy
| |
Collapse
|
123
|
Korkmaz OT, Ay H, Aytan N, Carreras I, Kowall NW, Dedeoglu A, Tuncel N. Vasoactive Intestinal Peptide Decreases β-Amyloid Accumulation and Prevents Brain Atrophy in the 5xFAD Mouse Model of Alzheimer's Disease. J Mol Neurosci 2018; 68:389-396. [PMID: 30498985 DOI: 10.1007/s12031-018-1226-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular deposits of fibrillary β-amyloid (Aβ) plaques in the brain that initiate an inflammatory process resulting in neurodegeneration. The neuronal loss associated with AD results in gross atrophy of affected regions causing a progressive loss of cognitive ability and memory function, ultimately leading to dementia. Growing evidence suggests that vasoactive intestinal peptide (VIP) could be beneficial for various neurodegenerative diseases, including AD. The study investigated the effects of VIP on 5xFAD, a transgenic mouse model of AD. Toward this aim, we used 20 5xFAD mice in two groups (n = 10 each), VIP-treated (25 ng/kg i.p. injection, three times per week) and saline-treated (the drug's vehicle) following the same administration regimen. Treatment started at 1 month of age and ended 2 months later. After 2 months of treatment, the mice were euthanized, their brains dissected out, and immunohistochemically stained for Aβ40 and Aβ42 on serial sections. Then, plaque analysis and stereological morphometric analysis were performed in different brain regions. Chronic VIP administration in 5xFAD mice significantly decreased the levels of Aβ40 and Aβ42 plaques in the subiculum compared to the saline treated 5xFAD mice. VIP treatment also significantly decreased Aβ40 and Aβ42 plaques in cortical areas and significantly increased the hippocampus/cerebrum and corpus callosum/cerebrum ratio but not the cerebral cortex/cerebrum ratio. In summary, we found that chronic administration of VIP significantly decreased Aβ plaques and preserved against atrophy for related brain regions in 5xFAD AD mice.
Collapse
Affiliation(s)
- Orhan Tansel Korkmaz
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA. .,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA. .,Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey. .,Faculty of Medicine, Department of Physiology, Eskisehir Osmangazi University, Odunpazari, 26040, Eskisehir, Turkey.
| | - Hakan Ay
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, 26040, Eskisehir, Turkey
| | - Nurgul Aytan
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Isabel Carreras
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Neil W Kowall
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Alpaslan Dedeoglu
- Research and Development, VA Boston Healthcare System, Boston, MA, 02130, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.,Department of Radiology, MGH and Harvard Medical School, Boston, MA, 02114, USA
| | - Nese Tuncel
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, 26480, Eskisehir, Turkey
| |
Collapse
|
124
|
Bisht K, Sharma K, Tremblay MÈ. Chronic stress as a risk factor for Alzheimer's disease: Roles of microglia-mediated synaptic remodeling, inflammation, and oxidative stress. Neurobiol Stress 2018; 9:9-21. [PMID: 29992181 PMCID: PMC6035903 DOI: 10.1016/j.ynstr.2018.05.003] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/23/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Microglia are the predominant immune cells of the central nervous system (CNS) that exert key physiological roles required for maintaining CNS homeostasis, notably in response to chronic stress, as well as mediating synaptic plasticity, learning and memory. The repeated exposure to stress confers a higher risk of developing neurodegenerative diseases including sporadic Alzheimer's disease (AD). While microglia have been causally linked to amyloid beta (Aβ) accumulation, tau pathology, neurodegeneration, and synaptic loss in AD, they were also attributed beneficial roles, notably in the phagocytic elimination of Aβ. In this review, we discuss the interactions between chronic stress and AD pathology, overview the roles played by microglia in AD, especially focusing on chronic stress as an environmental risk factor modulating their function, and present recently-described microglial phenotypes associated with neuroprotection in AD. These microglial phenotypes observed under both chronic stress and AD pathology may provide novel opportunities for the development of better-targeted therapeutic interventions.
Collapse
Key Words
- ABCA7, ATP-binding cassette transporter A7
- AD, Alzheimer's disease
- APOE, Apolipoprotein E
- APP, amyloid precursor protein
- Alzheimer's disease
- Aβ, Amyloid beta
- BDNF, brain derived neurotrophic factor
- CD11b, cluster of differentiation molecule 11B
- CD33, cluster of differentiation 33
- CNS, central nervous system
- CR, complement receptor
- CRF, corticotropin releasing factor
- DAM, disease associated microglia
- DAP12, DNAX-activation protein 12
- Dark microglia
- FAD, Familial Alzheimer's disease
- FCRLS, Fc receptor-like S scavenger receptor
- GR, glucocorticoid receptor
- HPA axis, hypothalamic pituitary adrenocortical axis
- IBA1, ionized calcium-binding adapter molecule 1
- IL, interleukin
- LTP, long-term potentiation
- MGnD, microglia with a neurodegenerative phenotype
- MR, mineralocorticoid receptor
- Microglia
- Microglial phenotypes
- NADPH, nicotinamide adenine dinucleotide phosphate
- NFT, neurofibrillary tangles
- Neurodegeneration
- Neuroinflammation
- PS, presenilin
- ROS, reactive oxygen species
- Stress
- Synaptic remodeling
- TGFβ, transforming growth factor β
- TLR, Toll-like receptors
- TMEM119, transmembrane protein 119
- TNFα, tumor necrosis factor-α
- TREM2, triggering receptor expressed in myeloid cells 2
- TYROBP, TYRO protein tyrosine kinase binding protein
- mPFC, medial prefrontal cortex
Collapse
Affiliation(s)
- Kanchan Bisht
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Kaushik Sharma
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
- Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
125
|
Rodriguez-Zas SL, Wu C, Southey BR, O'Connor JC, Nixon SE, Garcia R, Zavala C, Lawson M, McCusker RH, Romanova EV, Sweedler JV, Kelley KW, Dantzer R. Disruption of microglia histone acetylation and protein pathways in mice exhibiting inflammation-associated depression-like symptoms. Psychoneuroendocrinology 2018; 97:47-58. [PMID: 30005281 PMCID: PMC6138522 DOI: 10.1016/j.psyneuen.2018.06.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/22/2018] [Accepted: 06/29/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND Peripheral immune challenge can elicit microglia activation and depression-related symptoms. The balance of inflammatory signals in the tryptophan pathway can skew the activity of indoleamine-pyrrole 2,3 dioxygenase (IDO1) towards the metabolization of tryptophan into kynurenine (rather than serotonin), and towards neuroprotective or neurotoxic metabolites. The proteome changes that accompany inflammation-associated depression-related behaviors are incompletely understood. METHODS The changes in microglia protein abundance and post-translational modifications in wild type (WT) mice that exhibit depression-like symptoms after recovery from peripheral Bacille Calmette-Guerin (BCG) challenge were studied. This WT_BGG group was compared to mice that do not express depression-like symptoms after BCG challenge due to IDO1 deficiency by means of genetic knockout (BCG_KO group), and to WT Saline-treated (Sal) mice (WT_Sal group) using a mass spectrometry-based label-free approach. RESULTS The comparison of WT_BCG relative to WT_Sal and KO_BCG mice uncovered patterns of protein abundance and acetylation among the histone families that could influence microglia signaling and transcriptional rates. Members of the histone clusters 1, 2 and 3 families were less abundant in WT_BCG relative to WT_Sal whereas members in the H2A family exhibited the opposite pattern. Irrespective of family, the majority of the histones were less abundant in WT_BCG relative to KO_BCG microglia. Homeostatic mechanisms may temper the potentially toxic effects of high histone levels after BCG challenge to levels lower than Sal. Histone acetylation was highest in WT_BCG and the similar levels observed in WT_Sal and KO_BCG. This result suggest that histone acetylation levels are similar between IDO1 deficient mice after immune challenge and unchallenged WT mice. The over-abundance of tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation proteins (14-3-3 series) in WT_BCG relative to KO_BCG is particularly interesting because these proteins activate another rate-limiting enzyme in the tryptophan pathway. The over-representation of alcoholism and systemic lupus erythematosus pathways among the proteins exhibiting differential abundance between the groups suggest that these disorders share microglia activation pathways with BCG challenge. The over-representation of phagosome pathway among proteins differentially abundant between WT_BCG and KO_BCG microglia suggest an association between IDO1 deficiency and phagocytosis. Likewise, the over-representation of the gap junction pathway among the differentially abundant proteins between KO_BCG and WT_Sal suggest a multifactorial effect of BCG and IDO1 deficiency on cell communication. CONCLUSIONS The present study of histone acetylation and differential protein abundance furthers the understanding of the long lasting effects of peripheral immune challenges. Our findings offer insights into target proteins and mechanisms that provide clues for therapies to ameliorate inflammation-associated depression-related behaviors.
Collapse
Affiliation(s)
- Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Cong Wu
- Department of Biochemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jason C O'Connor
- Department of Pharmacology, University of Texas Health San Antonio and Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, TX, USA
| | - Scott E Nixon
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Robmay Garcia
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Cynthia Zavala
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marcus Lawson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Robert H McCusker
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Keith W Kelley
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
126
|
Qian L, Milne MR, Shepheard S, Rogers ML, Medeiros R, Coulson EJ. Removal of p75 Neurotrophin Receptor Expression from Cholinergic Basal Forebrain Neurons Reduces Amyloid-β Plaque Deposition and Cognitive Impairment in Aged APP/PS1 Mice. Mol Neurobiol 2018; 56:4639-4652. [DOI: 10.1007/s12035-018-1404-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
|
127
|
Abstract
Dietary and supplemental intake of the ω-3 fatty acid docosahexaenoic acid (DHA) reduces risk of Alzheimer’s disease (AD) and ameliorates symptoms. The apolipoprotein E (APOE)4 allele is the strongest risk factor for sporadic AD, exclusive of age. APOE4 carriers respond well to the DHA present in fish but do not respond as well to dietary supplements. The mechanisms behind this varied response remain unknown. I posit that the difference is that fish contain DHA in phospholipid form, whereas fish oil supplements do not. This influences whether DHA is metabolized to nonesterified DHA (free DHA) or a phospholipid form called lysophosphatidylcholine DHA (DHA-lysoPC). Free DHA is transported across the outer membrane leaflet of the blood–brain barrier (BBB) via passive diffusion, and DHA-lysoPC is transported across the inner membrane leaflet of the BBB via the major facilitator superfamily domain-containing protein 2A. I propose that APOE4 carriers have impaired brain transport of free DHA but not of DHA-lysoPC, as a consequence of a breakdown in the outer membrane leaflet of the BBB, putting them at increased risk for AD. Dietary sources of DHA in phospholipid form may provide a means to increase plasma levels of DHA-lysoPC, thereby decreasing the risk of AD.—Patrick, R. P. Role of phosphatidylcholine-DHA in preventing APOE4-associated Alzheimer’s disease.
Collapse
Affiliation(s)
- Rhonda P Patrick
- University of California San Francisco Benioff, Children's Hospital Oakland Research Institute, Oakland, California, USA
| |
Collapse
|
128
|
Lee JY, Joo B, Nam JH, Nam HY, Lee W, Nam Y, Seo Y, Kang HJ, Cho HJ, Jang YP, Kim J, We YM, Koo JW, Hoe HS. An Aqueous Extract of Herbal Medicine ALWPs Enhances Cognitive Performance and Inhibits LPS-Induced Neuroinflammation via FAK/NF-κB Signaling Pathways. Front Aging Neurosci 2018; 10:269. [PMID: 30319390 PMCID: PMC6168635 DOI: 10.3389/fnagi.2018.00269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that Liuwei Dihuang pills (LWPs) can positively affect learning, memory and neurogenesis. However, the underlying molecular mechanisms are not understood. In the present study, we developed ALWPs, a mixture of Antler and LWPs, and investigated whether ALWPs can affect neuroinflammatory responses. We found that ALWPs (500 mg/ml) inhibited lipopolysaccharide (LPS)-induced proinflammatory cytokine IL-1β mRNA levels in BV2 microglial cells but not primary astrocytes. ALWPs significantly reduced LPS-induced cell-surface levels of TLR4 to alter neuroinflammation. An examination of the molecular mechanisms by which ALWPs regulate the LPS-induced proinflammatory response revealed that ALWPs significantly downregulated LPS-induced levels of FAK phosphorylation, suggesting that ALWPs modulate FAK signaling to alter LPS-induced IL-1β levels. In addition, treatment with ALWPs followed by LPS resulted in decreased levels of the transcription factor NF-κB in the nucleus compared with LPS alone. Moreover, ALWPs significantly suppressed LPS-induced BV2 microglial cell migration. To examine whether ALWPs modulate learning and memory in vivo, wild-type C57BL/6J mice were orally administered ALWPs (200 mg/kg) or PBS daily for 3 days, intraperitoneally injected (i.p.) with LPS (250 μg/kg) or PBS, and assessed in Y maze and NOR tests. We observed that oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly rescued short- and long-term memory. More importantly, oral administration of ALWPs to LPS-injected wild-type C57BL/6J mice significantly reduced microglial activation in the hippocampus and cortex. Taken together, our results suggest that ALWPs can suppress neuroinflammation-associated cognitive deficits and that ALWPs have potential as a drug for neuroinflammation/neurodegeneration-related diseases, including Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Ju-Young Lee
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Bitna Joo
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Hye Yeon Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Wonil Lee
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Youngpyo Nam
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Yongtaek Seo
- Division of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Hye-Jin Kang
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Hyun-Ji Cho
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Young Pyo Jang
- Division of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, South Korea
| | - Jeongyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| | - Young-Man We
- College of Korean Medicine, Wonkwang University, Iksan, South Korea
- Oriental Medical Clinic Center, Hyoo Medical Clinic, Seoul, South Korea
| | - Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, South Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
129
|
Effect of growth differentiation factor-15 secreted by human umbilical cord blood-derived mesenchymal stem cells on amyloid beta levels in in vitro and in vivo models of Alzheimer's disease. Biochem Biophys Res Commun 2018; 504:933-940. [PMID: 30224067 DOI: 10.1016/j.bbrc.2018.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD), which is the most common progressive neurodegenerative disease, causes learning and memory impairment. The pathological progress of AD can derive from imbalanced homeostasis of amyloid beta (Aβ) in the brain. In such cases, microglia play important roles in regulating the brain Aβ levels. In the present study, we found that human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) can increase, through paracrine action, the ability of microglial cells to clear Aβ. In order to identify the associated paracrine factors, a secretome of hUCB-MSCs co-cultured with Aβ-treated BV2 microglial cells was analyzed using a human cytokine protein array. As a result, growth differentiation factor-15 (GDF-15) was identified as a predominant candidate, and its association with Aβ clearance by microglial cells was investigated in vitro and in a 5XFAD mouse model. When Aβ-treated BV2 cells were treated with exogenous recombinant GDF-15, the Aβ levels in the culture medium decreased. Moreover, GDF-15 injection in the brain parenchyma of 5XFAD mice also led to decrease in Aβ plaques. In contrast, co-culture of BV2 cells and hUCB-MSCs treated with GDF-15-specific siRNA did not influence the Aβ levels in the culture medium. To elucidate how these phenomena are related, we confirmed that GDF-15 specifically increases insulin-degrading enzyme (IDE) expression in microglial cells through TGFβ receptor type II (TGFβRII), both in vitro and in vivo. These findings suggest that hUCB-MSCs promote the Aβ clearance ability of microglial cells through regulation of GDF-15 secretion, thus elucidating a therapeutic mechanism for AD.
Collapse
|
130
|
Sejimo S, Hossain MS, Akashi K. Scallop-derived plasmalogens attenuate the activation of PKCδ associated with the brain inflammation. Biochem Biophys Res Commun 2018; 503:837-842. [DOI: 10.1016/j.bbrc.2018.06.084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 06/16/2018] [Indexed: 12/20/2022]
|
131
|
Herman F, Westfall S, Brathwaite J, Pasinetti GM. Suppression of Presymptomatic Oxidative Stress and Inflammation in Neurodegeneration by Grape-Derived Polyphenols. Front Pharmacol 2018; 9:867. [PMID: 30210334 PMCID: PMC6122113 DOI: 10.3389/fphar.2018.00867] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/17/2018] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative disorders constitute a group of multifaceted conditions characterized by the progressive loss of neurons and synaptic connections consequent to a combination of specific genetic predispositions and stochastic stressors. The neuropathologies observed in both Alzheimer's and Parkinson's disease are in part attributed to compounding intrinsic and extrinsic environmental stressors, which we propose may be limited by the administration of specific grape derived phytochemicals and their metabolized derivatives, specifically polyphenols isolated from grape botanicals. Current therapies for neurodegenerative disorders are limited as they solely target the final disease pathologies including behavioral changes, cognitive deficits, proteinopathies and neuronal loss; however, this strategy is not a sustainable approach toward managing disease onset or progression. This review discusses the application of grape derived polyphenols as an adjunctive treatment paradigm for the prevention of neuropathologies associated with Alzheimer's disease, Parkinson's disease and Chronic Traumatic Encephalopathy by simultaneously ameliorating two stochastic stressors that facilitate their disease pathologies: inflammation and oxidative stress. The biophysical attributes of grape-derived polyphenols buffer against redox potential dependent peripheral and neuroinflammation and down regulate the activation of inflammasomes in microglia and astrocytes, which could provide a novel mechanism through which grape-derived polyphenols simultaneously suppress risk factors across pathologically distinct neurodegenerative conditions. This approach therefore offers a prophylactic mode, not feasible through current pharmacological agents, to target activity dependent risk factors for neurodegenerative disorders that manifest over an individual's lifetime.
Collapse
Affiliation(s)
- Francis Herman
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
- Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY, United States
| | - Susan Westfall
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
- Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY, United States
| | - Justin Brathwaite
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
- Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY, United States
| | - Giulio M. Pasinetti
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
- Department of Genomic Sciences, Mount Sinai School of Medicine, New York, NY, United States
- James J. Peters VA Medical Center, Bronx, NY, United States
| |
Collapse
|
132
|
Ranjan VD, Qiu L, Tan EK, Zeng L, Zhang Y. Modelling Alzheimer's disease: Insights from in vivo to in vitro three-dimensional culture platforms. J Tissue Eng Regen Med 2018; 12:1944-1958. [PMID: 30011422 DOI: 10.1002/term.2728] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/21/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by progressive memory loss, impairment of other cognitive functions, and inability to perform activities of daily life. The key to understanding AD aetiology lies in the development of effective disease models, which should ideally recapitulate all aspects pertaining to the disease. A plethora of techniques including in vivo, in vitro, and in silico platforms have been utilized in developing disease models of AD over the years. Each of these approaches has revealed certain essential characteristics of AD; however, none have managed to fully mimic the pathological hallmarks observed in the AD human brain. In this review, we will provide details into the genesis, evolution, and significance of the principal methods currently employed in modelling AD, the advantages and limitations faced in their application, including the headways made by each approach. This review will focus primarily on two-dimensional and three-dimensional in vitro modelling of AD, which during the last few years has made significant breakthroughs in the areas of AD pathology and therapeutic screening. In addition, a glimpse into state-of-the-art neural tissue engineering techniques incorporating biomaterials and microfluidics technologies is provided, which could pave the way for the development of more accurate and comprehensive AD models in the future.
Collapse
Affiliation(s)
- Vivek Damodar Ranjan
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore.,School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore.,Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Yilei Zhang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
133
|
Zhu M, Wang X, Sun L, Schultzberg M, Hjorth E. Can inflammation be resolved in Alzheimer's disease? Ther Adv Neurol Disord 2018; 11:1756286418791107. [PMID: 30116300 PMCID: PMC6088473 DOI: 10.1177/1756286418791107] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 05/24/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive memory loss and dementia. Accumulating evidence suggests that inflammation is involved in the pathogenesis of AD. Epidemiological studies suggest that use of anti-inflammatory drugs is associated with a lower incidence of AD. However, clinical trials with anti-inflammatory drugs have not been successful. Recent studies have shown that inflammation is resolved by a process that is mediated by a group of lipid mediators, so called specialized pro-resolving lipid mediators (SPMs). Unlike anti-inflammatory strategies, which usually involve inhibition of the synthesis of inflammatory mediators, stimulating the resolution of inflammation is aimed at ending inflammation in a similar fashion as under normal physiological conditions. We have previously shown that pathways of resolution are impaired in AD. Moreover, we found that SPMs can improve neuronal survival and increase microglial phagocytosis of amyloid beta (Aβ) in in vitro studies, indicating that stimulating resolution of inflammation may be a potential therapeutic target in AD. In this review, we summarize recent findings regarding resolution of inflammation in AD. We also discuss possible strategies to stimulate the resolution of inflammation in AD, specifically focusing on signaling pathways, including SPMs, their receptors and enzymes involved in their formation.
Collapse
Affiliation(s)
- Mingqin Zhu
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Xinmin Street No 71, Changchun 130000, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, First Hospital of Jilin University, Changchun, China
| | - Marianne Schultzberg
- Department of Neurobiology, Care Sciences & Society, Section of Neurodegeneration, Karolinska Institutet, Center for Alzheimer Research, Sweden
| | - Erik Hjorth
- Department of Neurobiology, Care Sciences & Society, Section of Neurodegeneration, Karolinska Institutet, Center for Alzheimer Research, Sweden
| |
Collapse
|
134
|
Edison P, Donat CK, Sastre M. In vivo Imaging of Glial Activation in Alzheimer's Disease. Front Neurol 2018; 9:625. [PMID: 30131755 PMCID: PMC6090997 DOI: 10.3389/fneur.2018.00625] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by memory loss and decline of cognitive function, associated with progressive neurodegeneration. While neuropathological processes like amyloid plaques and tau neurofibrillary tangles have been linked to neuronal death in AD, the precise role of glial activation on disease progression is still debated. It was suggested that neuroinflammation could occur well ahead of amyloid deposition and may be responsible for clearing amyloid, having a neuroprotective effect; however, later in the disease, glial activation could become deleterious, contributing to neuronal toxicity. Recent genetic and preclinical studies suggest that the different activation states of microglia and astrocytes are complex, not as polarized as previously thought, and that the heterogeneity in their phenotype can switch during disease progression. In the last few years, novel imaging techniques e.g., new radiotracers for assessing glia activation using positron emission tomography and advanced magnetic resonance imaging technologies have emerged, allowing the correlation of neuro-inflammatory markers with cognitive decline, brain function and brain pathology in vivo. Here we review all new imaging technology in AD patients and animal models that has the potential to serve for early diagnosis of the disease, to monitor disease progression and to test the efficacy and the most effective time window for potential anti-inflammatory treatments.
Collapse
Affiliation(s)
- Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Cornelius K Donat
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| | - Magdalena Sastre
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
135
|
Model Senescent Microglia Induce Disease Related Changes in α-Synuclein Expression and Activity. Biomolecules 2018; 8:biom8030067. [PMID: 30071596 PMCID: PMC6164966 DOI: 10.3390/biom8030067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022] Open
Abstract
Aging is the most prominent risk factor for most neurodegenerative diseases. However, incorporating aging-related changes into models of neurodegeneration rarely occurs. One of the significant changes that occurs in the brain as we age is the shift in phenotype of the resident microglia population to one less able to respond to deleterious changes in the brain. These microglia are termed dystrophic microglia. In order to better model neurodegenerative diseases, we have developed a method to convert microglia into a senescent phenotype in vitro. Mouse microglia grown in high iron concentrations showed many characteristics of dystrophic microglia including, increased iron storage, increased expression of proteins, such as ferritin and the potassium channel, Kv1.3, increased reactive oxygen species production and cytokine release. We have applied this new model to the study of α-synuclein, a protein that is closely associated with a number of neurodegenerative diseases. We have shown that conditioned medium from our model dystrophic microglia increases α-synuclein transcription and expression via tumor necrosis factor alpha (TNFα) and mediated through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The conditioned medium also decreases the formation of α-synuclein tetramers, associated ferrireductase activity, and increases aggregates of α-synuclein. The results suggest that we have developed an interesting new model of aged microglia and that factors, including TNFα released from dystrophic microglia could have a significant influence on the pathogenesis of α-synuclein related diseases.
Collapse
|
136
|
Cobourne-Duval MK, Taka E, Mendonca P, Soliman KFA. Thymoquinone increases the expression of neuroprotective proteins while decreasing the expression of pro-inflammatory cytokines and the gene expression NFκB pathway signaling targets in LPS/IFNγ -activated BV-2 microglia cells. J Neuroimmunol 2018; 320:87-97. [PMID: 29759145 PMCID: PMC5967628 DOI: 10.1016/j.jneuroim.2018.04.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/17/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
Abstract
Neuroinflammation and microglial activation are pathological markers of a number of central nervous system (CNS) diseases. Chronic activation of microglia induces the release of excessive amounts of reactive oxygen species (ROS) and pro-inflammatory cytokines. Additionally, chronic microglial activation has been implicated in several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Thymoquinone (TQ) has been identified as one of the major active components of the natural product Nigella sativa seed oil. TQ has been shown to exhibit anti-inflammatory, anti-oxidative, and neuroprotective effects. In this study, lipopolysaccharide (LPS) and interferon gamma (IFNγ) activated BV-2 microglial cells were treated with TQ (12.5 μM for 24 h). We performed quantitative proteomic analysis using Orbitrap/Q-Exactive Proteomic LC-MS/MS (Liquid chromatography-mass spectrometry) to globally assess changes in protein expression between the treatment groups. Furthermore, we evaluated the ability of TQ to suppress the inflammatory response using ELISArray™ for Inflammatory Cytokines. We also assessed TQ's effect on the gene expression of NFκB signaling targets by profiling 84 key genes via real-time reverse transcription (RT2) PCR array. Our results indicated that TQ treatment of LPS/IFNγ-activated microglial cells significantly increased the expression of 4 antioxidant, neuroprotective proteins: glutaredoxin-3 (21 fold; p < 0.001), biliverdin reductase A (15 fold; p < 0.0001), 3-mercaptopyruvate sulfurtransferase (11 fold; p < 0.01), and mitochondrial lon protease (>8 fold; p < 0.001) compared to the untreated, activated cells. Furthermore, TQ treatment significantly (P < 0.0001) reduced the expression of inflammatory cytokines, IL-2 = 38%, IL-4 = 19%, IL-6 = 83%, IL-10 = 237%, and IL-17a = 29%, in the activated microglia compared to the untreated, activated which expression levels were significantly elevated compared to the control microglia: IL-2 = 127%, IL-4 = 151%, IL-6 = 670%, IL-10 = 133%, IL-17a = 127%. Upon assessing the gene expression of NFκB signaling targets, this study also demonstrated that TQ treatment of activated microglia resulted in >7 fold down-regulation of several NFκB signaling targets genes, including interleukin 6 (IL6), complement factor B (CFB), chemokine (CC motif) ligand 3 (CXCL3), chemokine (CC) motif ligand 5 (CCL5) compared to the untreated, activated microglia. This modulation in gene expression counteracts the >10-fold upregulation of these same genes observed in the activated microglia compared to the controls. Our results show that TQ treatment of LPS/IFNγ-activated BV-2 microglial cells induce a significant increase in expression of neuroprotective proteins, a significant decrease in expression inflammatory cytokines, and a decrease in the expression of signaling target genes of the NFκB pathway. Our findings are the first to show that TQ treatment increased the expression of these neuroprotective proteins (biliverdin reductase-A, 3-mercaptopyruvate sulfurtransferase, glutaredoxin-3, and mitochondrial lon protease) in the activated BV-2 microglial cells. Additionally, our results indicate that TQ treatment decreased the activation of the NFκB signaling pathway, which plays a key role in neuroinflammation. In conclusion, our results demonstrate that TQ treatment reduces the inflammatory response and modulates the expression of specific proteins and genes and hence potentially reduce neuroinflammation and neurodegeneration driven by microglial activation.
Collapse
Affiliation(s)
- Makini K Cobourne-Duval
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States
| | - Equar Taka
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States
| | - Patricia Mendonca
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States
| | - Karam F A Soliman
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, United States.
| |
Collapse
|
137
|
Abstract
SIGNIFICANCE The p53 family of transcription factors, including p53, p63, and p73, plays key roles in both biological and pathological processes, including cancer and neural development. Recent Advances: In recent years, a growing body of evidence has indicated that the entire p53 family is involved in the regulation of the central nervous system (CNS) functions as well as in the pathogenesis of several neurological disorders. Mechanistically, the p53 proteins control neuronal cell fate, terminal differentiation, and survival, via a complex interplay among the family members. CRITICAL ISSUES In this article, we discuss the involvement of the p53 family in neurobiology and in pathological conditions affecting the CNS, including neuroinflammation. FUTURE DIRECTIONS Understanding the molecular mechanism(s) underlying the function of the p53 family could improve our general knowledge of the pathogenesis of brain disorders and potentially pave the road for new therapeutic intervention. Antioxid. Redox Signal. 29, 1-14.
Collapse
Affiliation(s)
- Massimiliano Agostini
- 1 Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata," Rome, Italy .,2 Medical Research Council, Toxicology Unit, Leicester University , Leicester, United Kingdom
| | - Gerry Melino
- 1 Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata," Rome, Italy .,2 Medical Research Council, Toxicology Unit, Leicester University , Leicester, United Kingdom
| | - Francesca Bernassola
- 1 Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata," Rome, Italy
| |
Collapse
|
138
|
Chatterjee S, Mudher A. Alzheimer's Disease and Type 2 Diabetes: A Critical Assessment of the Shared Pathological Traits. Front Neurosci 2018; 12:383. [PMID: 29950970 PMCID: PMC6008657 DOI: 10.3389/fnins.2018.00383] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) and Type 2 Diabetes Mellitus (T2DM) are two of the most prevalent diseases in the elderly population worldwide. A growing body of epidemiological studies suggest that people with T2DM are at a higher risk of developing AD. Likewise, AD brains are less capable of glucose uptake from the surroundings resembling a condition of brain insulin resistance. Pathologically AD is characterized by extracellular plaques of Aβ and intracellular neurofibrillary tangles of hyperphosphorylated tau. T2DM, on the other hand is a metabolic disorder characterized by hyperglycemia and insulin resistance. In this review we have discussed how Insulin resistance in T2DM directly exacerbates Aβ and tau pathologies and elucidated the pathophysiological traits of synaptic dysfunction, inflammation, and autophagic impairments that are common to both diseases and indirectly impact Aβ and tau functions in the neurons. Elucidation of the underlying pathways that connect these two diseases will be immensely valuable for designing novel drug targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Shreyasi Chatterjee
- Centre of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Amritpal Mudher
- Centre of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
139
|
Wang J, Xing H, Wan L, Jiang X, Wang C, Wu Y. Treatment targets for M2 microglia polarization in ischemic stroke. Biomed Pharmacother 2018; 105:518-525. [PMID: 29883947 DOI: 10.1016/j.biopha.2018.05.143] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023] Open
Abstract
As the first line of defense in the nervous system, resident microglia are the predominant immune cells in the brain. In diseases of the central nervous system such as stroke, Alzheimer's disease, and Parkinson's disease, they often cause inflammation or phagocytosis; however, some studies have found that despite the current controversy over M1, M2 polarization could be beneficial. Ischemic stroke is the third most common cause of death in humans. Patients who survive an ischemic stroke might experience a clear decline in their quality of life, owing to conditions such as hemiplegic paralysis and aphasia. After stroke, the activated microglia become a double-edged sword, with distinct phenotypic changes to the deleterious M1 and neuroprotective M2 types. Therefore, methods for promoting the differentiation of microglia into the M2 polarized form to alleviate harmful reactions after stroke have become a topic of interest in recent years. Subsequently, the discovery of new drugs related to M2 polarization has enabled the realization of targeted therapies. In the present review, we discussed the neuroprotective effects of microglia M2 polarization and the potential mechanisms and drugs by which microglia can be transformed into the M2 polarized type after stroke.
Collapse
Affiliation(s)
- Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongyi Xing
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Wan
- The Children's Hospital of Soochow, Jiangsu, Hematology and Oncology, China
| | - Xingjun Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
140
|
Pantaleão L, Rocha GHO, Reutelingsperger C, Tiago M, Maria-Engler SS, Solito E, Farsky SP. Connections of annexin A1 and translocator protein-18 kDa on toll like receptor stimulated BV-2 cells. Exp Cell Res 2018; 367:282-290. [DOI: 10.1016/j.yexcr.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/06/2018] [Accepted: 04/07/2018] [Indexed: 01/27/2023]
|
141
|
Sarkar S, Malovic E, Sarda D, Lawana V, Rokad D, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Characterization and comparative analysis of a new mouse microglial cell model for studying neuroinflammatory mechanisms during neurotoxic insults. Neurotoxicology 2018; 67:129-140. [PMID: 29775624 DOI: 10.1016/j.neuro.2018.05.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
Microglia are the first responders of the central nervous system, acting as the key modulators of neuroinflammation observed during neurotoxic insults as well as in the pathophysiology of several neurodegenerative disorders including Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD). The number of publications on microglia has increased steadily throughout the past decade because of immense interests in the neuroinflammation that precedes the neurodegenerative process. To study microglial biology and its role in modulating neuroinflammation, immortalized microglial cell lines derived from mice, rats, and humans have been developed. Among these, the BV2 mouse microglial cell line is the most well characterized and widely used cell culture model. However, even unstimulated BV2 cells exhibit an amoeboid, hypertrophied morphology, indicating a highly activated and inflammatory state compared to primary microglia, thus making them less than ideal for studying the low-dose effects of toxicants on microglial activation. Therefore, we performed an in-depth characterization of a recently developed mouse microglial cell (MMC) line, which we compared with primary mouse microglia (PMG) and BV2s to identify which cell line was best suited for studying the microglial response to neurotoxicants. Comparative analyses reveal that MMCs are strikingly more similar to PMGs in basal activity, morphology, and sensitivity, than are BV2s. Furthermore, basal nitrite and inflammatory cytokine levels are significantly higher in BV2s compared to MMCs. BV2 cells are also less reactive to the inflammagen LPS compared to MMCs, due to the higher basal activation state of BV2s. Collectively, our in-depth analyses of morphology, basal activity, and responsivity to two different stimuli (LPS, aggregated α-synuclein) demonstrate that MMCs closely mimic neonatal PMGs, and are discernibly more suitable than BV2s for studying the neuroinflammatory mechanisms of neurotoxicants.
Collapse
Affiliation(s)
- Souvarish Sarkar
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Emir Malovic
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Deeksha Sarda
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Vivek Lawana
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Dharmin Rokad
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Huajun Jin
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Vellareddy Anantharam
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Arthi Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA, 50011, United States.
| |
Collapse
|
142
|
Training Microglia to Resist Alzheimer's Disease. J Neurosci 2018; 37:477-479. [PMID: 28100733 DOI: 10.1523/jneurosci.3345-16.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022] Open
|
143
|
Tang M, Taghibiglou C. The Mechanisms of Action of Curcumin in Alzheimer's Disease. J Alzheimers Dis 2018; 58:1003-1016. [PMID: 28527218 DOI: 10.3233/jad-170188] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of the elderly. As the prevalence of AD rises in the 21st century, there is an urgent need for the development of effective pharmacotherapies. Currently, drug treatments target the symptoms of the disease and do not modify or halt the disease progress. Thus, natural compounds have been investigated for their ability to treat AD. This review examines the efficacy of curcumin, a polyphenol derived from turmeric herb, to treat AD. We summarize the in vivo and in vitro research describing the mechanisms of action in which curcumin modifies AD pathology: curcumin inhibits the formation and promotes the disaggregation of amyloid-β plaques, attenuates the hyperphosphorylation of tau and enhances its clearance, binds copper, lowers cholesterol, modifies microglial activity, inhibits acetylcholinesterase, mediates the insulin signaling pathway, and is an antioxidant. In conclusion, curcumin has the potential to be more efficacious than current treatments. However, its usefulness as a therapeutic agent may be hindered by its low bioavailability. If the challenge of low bioavailability is overcome, curcumin-based medications for AD may be in the horizon.
Collapse
Affiliation(s)
- Mengxi Tang
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Changiz Taghibiglou
- Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
144
|
Lee AY, Lee MH, Lee S, Cho EJ. Neuroprotective Effect of Alpha-Linolenic Acid against Aβ-Mediated Inflammatory Responses in C6 Glial Cell. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4853-4861. [PMID: 29668263 DOI: 10.1021/acs.jafc.8b00836] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Therapeutic approaches for neurodegeneration, such as Alzheimer's disease (AD), have been widely studied. One of the critical hallmarks of AD is accumulation of amyloid beta (Aβ). Aβ induces neurotoxicity and releases inflammatory mediators or cytokines through activation of glial cell, and these pathological features are observed in AD patient's brain. The purpose of this study is to investigate the protective effect of alpha-linolenic acid (ALA) on Aβ25-35-induced neurotoxicity in C6 glial cells. Exposure of C6 glial cells to 50 μM Aβ25-35 caused cell death, overproduction of nitric oxide (NO), and pro-inflammatory cytokines release [interleukin (IL)-6 and tumor necrosis factor-α], while treatment of ALA increased cell viability and markedly attenuated Aβ25-35-induced excessive production of NO and those inflammatory cytokines. Inhibitory effect of ALA on generation of NO and cytokines was mediated by down-regulation of inducible nitric oxide synthase and cyclooxygenase-2 protein and mRNA expressions. In addition, ALA treatment inhibited reactive oxygen species generation induced by Aβ25-35 through the enhancement of the nuclear factor-erythroid 2-related factor-2 (Nrf-2) protein levels and subsequent induction of heme-oxygenase-1 (HO-1) expression in C6 glial cells dose- and time-dependently. Furthermore, the levels of neprilysin and insulin-degrading enzyme protein expressions, which contribute to degradation of Aβ, were also increased by treatment of ALA compared to Aβ25-35-treated control group. In conclusion, effects of ALA on Aβ degradation were shown to be mediated through inhibition of inflammatory responses and activation of antioxidative system, Nrf-2/HO-1 signaling pathway, in C6 glial cells. Our findings suggest that ALA might have the potential for therapeutics of AD.
Collapse
Affiliation(s)
- Ah Young Lee
- Department of Food Science and Nutrition & Kimchi Research Institute , Pusan National University , Busan 46241 , Republic of Korea
| | - Myoung Hee Lee
- Department of Southern Area Crop Science , National Institute of Crop Science, Rural Development Administration , Gyeongnam 50424 , Republic of Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science , Chung-Ang University , Gyeonggi 17546 , Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute , Pusan National University , Busan 46241 , Republic of Korea
| |
Collapse
|
145
|
Abstract
Alzheimer's disease (AD) is a debilitating disease influencing a multitude of outcomes, including memory function. Recent work suggests that memory may be influenced by exercise ('memorcise'), even among those with AD. The present narrative review details (1) the underlying mechanisms of AD; (2) whether exercise has a protective effect in preventing AD; (3) the mechanisms through which exercise may help to prevent AD; (4) the mechanisms through which exercise may help attenuate the progression of AD severity among those with existing AD; (5) the effects and mechanisms through which exercise is associated with memory among those with existing AD; and (6) exercise recommendations for those with existing AD. Such an understanding will aid clinicians in their ability to use exercise as a potential behavioral strategy to help prevent and treat AD.
Collapse
Affiliation(s)
- Paul D Loprinzi
- a Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management , The University of Mississippi , University , MS , USA
| | - Emily Frith
- a Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management , The University of Mississippi , University , MS , USA
| | - Pamela Ponce
- a Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management , The University of Mississippi , University , MS , USA
| |
Collapse
|
146
|
Lancour D, Naj A, Mayeux R, Haines JL, Pericak-Vance MA, Schellenberg GD, Crovella M, Farrer LA, Kasif S. One for all and all for One: Improving replication of genetic studies through network diffusion. PLoS Genet 2018; 14:e1007306. [PMID: 29684019 PMCID: PMC5933817 DOI: 10.1371/journal.pgen.1007306] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 05/03/2018] [Accepted: 03/11/2018] [Indexed: 12/31/2022] Open
Abstract
Improving accuracy in genetic studies would greatly accelerate understanding the genetic basis of complex diseases. One approach to achieve such an improvement for risk variants identified by the genome wide association study (GWAS) approach is to incorporate previously known biology when screening variants across the genome. We developed a simple approach for improving the prioritization of candidate disease genes that incorporates a network diffusion of scores from known disease genes using a protein network and a novel integration with GWAS risk scores, and tested this approach on a large Alzheimer disease (AD) GWAS dataset. Using a statistical bootstrap approach, we cross-validated the method and for the first time showed that a network approach improves the expected replication rates in GWAS studies. Several novel AD genes were predicted including CR2, SHARPIN, and PTPN2. Our re-prioritized results are enriched for established known AD-associated biological pathways including inflammation, immune response, and metabolism, whereas standard non-prioritized results were not. Our findings support a strategy of considering network information when investigating genetic risk factors. Integrating multiple types of -omics data is a rapidly growing research area due in part to the increasing amount of diverse and publicly accessible data. In this study, we demonstrated that integration of genetic association and protein interaction data using a network diffusion approach measurably improves reproducibility of top candidate genes. Application of this approach to Alzheimer disease (AD) using a large dataset assembled by the Alzheimer’s Disease Genetics Consortium identified several novel candidate AD genes that are supported by pre-existing knowledge of AD pathobiology. Our findings support a strategy of considering network information when investigating genetic risk factors. Finally, we developed a transparent and easy-to-use R package that can facilitate the extension of our methodology to other phenotypes for which genetic data are available.
Collapse
Affiliation(s)
- Daniel Lancour
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Adam Naj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Richard Mayeux
- Department of Neurology and Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Margaret A. Pericak-Vance
- Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark Crovella
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Computer Science, Boston University, Boston, Massachusetts, United States of America
| | - Lindsay A. Farrer
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Medicine (Biomedical Genetics), Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, United States of America
- * E-mail:
| | - Simon Kasif
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| |
Collapse
|
147
|
Pfeffer A, Munder T, Schreyer S, Klein C, Rasińska J, Winter Y, Steiner B. Behavioral and psychological symptoms of dementia (BPSD) and impaired cognition reflect unsuccessful neuronal compensation in the pre-plaque stage and serve as early markers for Alzheimer's disease in the APP23 mouse model. Behav Brain Res 2018; 347:300-313. [PMID: 29572105 DOI: 10.1016/j.bbr.2018.03.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/28/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022]
Abstract
Recent research on Alzheimer's disease (AD) focuses on processes prior to amyloid-beta plaque deposition accounting for the progress of the disease. However, early mechanisms of AD are still poorly understood and predictors of the disease in the pre-plaque stage essential for initiating an early therapy are lacking. Behavioral and psychological symptoms of dementia (BPSD) and potentially impaired cognition may serve as predictors and early clinical diagnostic markers for AD. To investigate potential BPSD and cognitive impairments in association with neuronal cell development as such markers for AD in the pre-plaque stage, female APP23 mice at eight, 19 and 31 weeks of age and corresponding control animals were tested for BPSD (elevated zero maze; sucrose preference test), motor coordination (rotarod), spatial memory and reversal learning (Morris water maze) and hippocampal neurogenesis as a neuronal correlate for hippocampus-dependent behavior. To evaluate a potential therapeutic effect of physical, cognitive and social stimulation, animals were exposed to environmental enrichment (EE) for one, twelve or 24 weeks from five weeks of age. In APP23, decreased anxiety accompanied increased agitation from eight weeks of age. Impairment of spatial memory and learning flexibility prior to plaque deposition involved an insufficient use of spatial search strategies associated with an unsuccessful compensatory increase of neurogenesis. EE had an overall beneficial effect on behavior and neurogenesis and thus constitutes a therapeutic tool to slow disease progression. BPSD, cognition and associated impaired neurogenesis complement clinical diagnostic markers for pre-plaque AD and contribute to an early detection essential to halt disease progression.
Collapse
Affiliation(s)
- Anna Pfeffer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Tonia Munder
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefanie Schreyer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Charlotte Klein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Justyna Rasińska
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - York Winter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany
| | - Barbara Steiner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
148
|
The Role of Macrophages in Neuroinflammatory and Neurodegenerative Pathways of Alzheimer's Disease, Amyotrophic Lateral Sclerosis, and Multiple Sclerosis: Pathogenetic Cellular Effectors and Potential Therapeutic Targets. Int J Mol Sci 2018. [PMID: 29533975 PMCID: PMC5877692 DOI: 10.3390/ijms19030831] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In physiological conditions, different types of macrophages can be found within the central nervous system (CNS), i.e., microglia, meningeal macrophages, and perivascular (blood-brain barrier) and choroid plexus (blood-cerebrospinal fluid barrier) macrophages. Microglia and tissue-resident macrophages, as well as blood-borne monocytes, have different origins, as the former derive from yolk sac erythromyeloid precursors and the latter from the fetal liver or bone marrow. Accordingly, specific phenotypic patterns characterize each population. These cells function to maintain homeostasis and are directly involved in the development and resolution of neuroinflammatory processes. Also, following inflammation, circulating monocytes can be recruited and enter the CNS, therefore contributing to brain pathology. These cell populations have now been identified as key players in CNS pathology, including autoimmune diseases, such as multiple sclerosis, and degenerative diseases, such as Amyotrophic Lateral Sclerosis and Alzheimer’s disease. Here, we review the evidence on the involvement of CNS macrophages in neuroinflammation and the advantages, pitfalls, and translational opportunities of pharmacological interventions targeting these heterogeneous cellular populations for the treatment of brain diseases.
Collapse
|
149
|
Jha SK, Jha NK, Kumar D, Sharma R, Shrivastava A, Ambasta RK, Kumar P. Stress-Induced Synaptic Dysfunction and Neurotransmitter Release in Alzheimer's Disease: Can Neurotransmitters and Neuromodulators be Potential Therapeutic Targets? J Alzheimers Dis 2018; 57:1017-1039. [PMID: 27662312 DOI: 10.3233/jad-160623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The communication between neurons at synaptic junctions is an intriguing process that monitors the transmission of various electro-chemical signals in the central nervous system. Albeit any aberration in the mechanisms associated with transmission of these signals leads to loss of synaptic contacts in both the neocortex and hippocampus thereby causing insidious cognitive decline and memory dysfunction. Compelling evidence suggests that soluble amyloid-β (Aβ) and hyperphosphorylated tau serve as toxins in the dysfunction of synaptic plasticity and aberrant neurotransmitter (NT) release at synapses consequently causing a cognitive decline in Alzheimer's disease (AD). Further, an imbalance between excitatory and inhibitory neurotransmission systems induced by impaired redox signaling and altered mitochondrial integrity is also amenable for such abnormalities. Defective NT release at the synaptic junction causes several detrimental effects associated with altered activity of synaptic proteins, transcription factors, Ca2+ homeostasis, and other molecules critical for neuronal plasticity. These detrimental effects further disrupt the normal homeostasis of neuronal cells and thereby causing synaptic loss. Moreover, the precise mechanistic role played by impaired NTs and neuromodulators (NMs) and altered redox signaling in synaptic dysfunction remains mysterious, and their possible interlink still needs to be investigated. Therefore, this review elucidates the intricate role played by both defective NTs/NMs and altered redox signaling in synaptopathy. Further, the involvement of numerous pharmacological approaches to compensate neurotransmission imbalance has also been discussed, which may be considered as a potential therapeutic approach in synaptopathy associated with AD.
Collapse
|
150
|
Bastian FO. Combined Creutzfeldt-Jakob/ Alzheimer's Disease Cases are Important in Search for Microbes in Alzheimer's Disease. J Alzheimers Dis 2018; 56:867-873. [PMID: 28059790 DOI: 10.3233/jad-160999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The question whether Alzheimer's disease is infectious as brought up in the recent editorial published in the Journal of Alzheimer's Disease is complicated by the controversy whether the causal agent is a microbe or a misfolded host protein (amyloid). The replicating amyloid (prion) theory, based upon data from studies of Creutzfeldt-Jakob disease (CJD) and other transmissible spongiform encephalopathies (TSEs), has been challenged since the prion can be separated from TSE infectivity, and spiroplasma, a wall-less bacterium, has been shown to be involved in the pathogenesis of CJD. Further support for a microbial cause for AD comes from occurrence of mixed CJD/AD cases involving up to 15% of AD brains submitted to brain banks. The association of CJD with AD suggests a common etiology rather than simply being a medical curiosity. A co-infection with the transmissible agent of CJD, which we propose to be a Spiroplasma sp., would explain the diversity of bacteria shown to be associated with cases of AD.
Collapse
Affiliation(s)
- Frank O Bastian
- School of Animal Science, Louisiana State University Agricultural Center, Baton Rouge, LA, USA.,Tulane Medical School, New Orleans, LA, USA.,Texas Tech University, Lubbock, TX, USA
| |
Collapse
|