1
|
Huang L, Liu M, Li Z, Li B, Wang J, Zhang K. Systematic review of amyloid-beta clearance proteins from the brain to the periphery: implications for Alzheimer's disease diagnosis and therapeutic targets. Neural Regen Res 2025; 20:3574-3590. [PMID: 39820231 DOI: 10.4103/nrr.nrr-d-24-00865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025] Open
Abstract
Amyloid-beta clearance plays a key role in the pathogenesis of Alzheimer's disease. However, the variation in functional proteins involved in amyloid-beta clearance and their correlation with amyloid-beta levels remain unclear. In this study, we conducted meta-analyses and a systematic review using studies from the PubMed, Embase, Web of Science, and Cochrane Library databases, including journal articles published from inception to June 30, 2023. The inclusion criteria included studies comparing the levels of functional proteins associated with amyloid-beta clearance in the blood, cerebrospinal fluid, and brain of healthy controls, patients with mild cognitive impairment, and patients with Alzheimer's disease. Additionally, we analyzed the correlation between these functional proteins and amyloid-beta levels in patients with Alzheimer's disease. The methodological quality of the studies was assessed via the Newcastle‒Ottawa Scale. Owing to heterogeneity, we utilized either a fixed-effect or random-effect model to assess the 95% confidence interval (CI) of the standard mean difference (SMD) among healthy controls, patients with mild cognitive impairment, and patients with Alzheimer's disease. The findings revealed significant alterations in the levels of insulin-degrading enzymes, neprilysin, matrix metalloproteinase-9, cathepsin D, receptor for advanced glycation end products, and P-glycoprotein in the brains of patients with Alzheimer's disease, patients with mild cognitive impairment, and healthy controls. In cerebrospinal fluid, the levels of triggering receptor expressed on myeloid cells 2 and ubiquitin C-terminal hydrolase L1 are altered, whereas the levels of TREM2, CD40, CD40L, CD14, CD22, cathepsin D, cystatin C, and α2 M in peripheral blood differ. Notably, TREM2 and cathepsin D showed changes in both brain (SMD = 0.31, 95% CI: 0.16-0.47, P < 0.001, I2 = 78.4%; SMD = 1.24, 95% CI: 0.01-2.48, P = 0.048, I2 = 90.1%) and peripheral blood (SMD = 1.01, 95% CI: 0.35-1.66, P = 0.003, I2 = 96.5%; SMD = 7.55, 95% CI: 3.92-11.18, P < 0.001, I2 = 98.2%) samples. Furthermore, correlations were observed between amyloid-beta levels and the levels of TREM2 ( r = 0.16, 95% CI: 0.04-0.28, P = 0.009, I2 = 74.7%), neprilysin ( r = -0.47, 95% CI: -0.80-0.14, P = 0.005, I2 = 76.1%), and P-glycoprotein ( r = -0.31, 95% CI: -0.51-0.11, P = 0.002, I2 = 0.0%) in patients with Alzheimer's disease. These findings suggest that triggering receptor expressed on myeloid cells 2 and cathepsin D could serve as potential diagnostic biomarkers for Alzheimer's disease, whereas triggering receptor expressed on myeloid cells 2, neprilysin, and P-glycoprotein may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Letian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mingyue Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Ze Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Bing Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| | - Jiahe Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
2
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2025; 36:139-168. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
3
|
Liu JC, Lei SY, Zhang DH, He QY, Sun YY, Zhu HJ, Qu Y, Zhou SY, Yang Y, Li C, Guo ZN. The pleiotropic effects of statins: a comprehensive exploration of neurovascular unit modulation and blood-brain barrier protection. Mol Med 2024; 30:256. [PMID: 39707228 DOI: 10.1186/s10020-024-01025-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The blood-brain barrier (BBB) is the most central component of the neurovascular unit (NVU) and is crucial for the maintenance of the internal environment of the central nervous system and the regulation of homeostasis. A multitude of neuroprotective agents have been developed to exert neuroprotective effects and improve the prognosis of patients with ischemic stroke. These agents have been designed to maintain integrity and promote BBB repair. Statins are widely used as pharmacological agents for the treatment and prevention of ischemic stroke, making them a cornerstone in the pharmacological armamentarium for this condition. The primary mechanism of action is the reduction of serum cholesterol through the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which results in a decrease in low-density lipoprotein cholesterol (LDL-C) and an increase in cholesterol clearance. Nevertheless, basic and clinical research has indicated that statins may exert additional pleiotropic effects beyond LDL-C reduction. Previous studies on ischemic stroke have demonstrated that statins can enhance neurological function, reduce inflammation, and promote angiogenic and synaptic processes following ischemic stroke. The BBB has been increasingly recognized for its role in the development and progression of ischemic stroke. Statins have also been found to play a potential BBB protective role by affecting members of the NVU. This review aimed to provide a comprehensive theoretical basis for the clinical application of statins by systematically detailing how statins influence the BBB, particularly focusing on the regulation of the function of each member of the NVU.
Collapse
Affiliation(s)
- Jia-Cheng Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Shuang-Yin Lei
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Ying-Ying Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Hong-Jing Zhu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China
| | - Chao Li
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China.
- Neuroscience Research Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| |
Collapse
|
4
|
Kwon HJ, Santhosh D, Huang Z. A novel monomeric amyloid β-activated signaling pathway regulates brain development via inhibition of microglia. eLife 2024; 13:RP100446. [PMID: 39635981 PMCID: PMC11620749 DOI: 10.7554/elife.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Amyloid β (Aβ) forms aggregates in the Alzheimer's disease brain and is well known for its pathological roles. Recent studies show that it also regulates neuronal physiology in the healthy brain. Whether Aβ also regulates glial physiology in the normal brain, however, has remained unclear. In this article, we describe the discovery of a novel signaling pathway activated by the monomeric form of Aβ in vitro that plays essential roles in the regulation of microglial activity and the assembly of neocortex during mouse development in vivo. We find that activation of this pathway depends on the function of amyloid precursor and the heterotrimeric G protein regulator Ric8a in microglia and inhibits microglial immune activation at transcriptional and post-transcriptional levels. Genetic disruption of this pathway during neocortical development results in microglial dysregulation and excessive matrix proteinase activation, leading to basement membrane degradation, neuronal ectopia, and laminar disruption. These results uncover a previously unknown function of Aβ as a negative regulator of brain microglia and substantially elucidate the underlying molecular mechanisms. Considering the prominence of Aβ and neuroinflammation in the pathology of Alzheimer's disease, they also highlight a potentially overlooked role of Aβ monomer depletion in the development of the disease.
Collapse
Affiliation(s)
- Hyo Jun Kwon
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Devi Santhosh
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Zhen Huang
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
5
|
Tang J, Cao Z, Lei M, Yu Q, Mai Y, Xu J, Liao W, Ruan Y, Shi L, Yang L, Liu J. Heterogeneity of cerebral atrophic rate in mild cognitive impairment and its interactive association with proteins related to microglia activity on longitudinal cognitive changes. Arch Gerontol Geriatr 2024; 127:105582. [PMID: 39079281 DOI: 10.1016/j.archger.2024.105582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Heterogeneity of cerebral atrophic rate commonly exists in mild cognitive impairment (MCI), which may be associated with microglia-involved neuropathology and have an influence on cognitive outcomes. OBJECTIVE We aim to explore the heterogeneity of cerebral atrophic rate among MCI and its association with plasma proteins related to microglia activity, with further investigation of their interaction effects on long-term cognition. SUBJECTS A total of 630 MCI subjects in the ADNI database were included, of which 260 subjects were available with baseline data on plasma proteins. METHODS Group-based multi-trajectory modeling (GBMT) was used to identify the latent classes with heterogeneous cerebral atrophic rates. Associations between latent classes and plasma proteins related to microglia activity were investigated with generalized linear models. Linear mixed effect models (LME) were implemented to explore the interaction effects between proteins related to microglia activity and identified latent classes on longitudinal cognitive changes. RESULTS Two latent classes were identified and labeled as the slow-atrophy class and the fast-atrophy class. Associations were found between such heterogeneity of atrophic rates and plasma proteins related to microglia activity, especially AXL receptor tyrosine kinase (AXL), CD40 antigen (CD40), and tumor necrosis factor receptor-like 2 (TNF-R2). Interaction effects on longitudinal cognitive changes showed that higher CD40 was associated with faster cognitive decline in the slow-atrophy class and higher AXL or TNF-R2 was associated with slower cognitive decline in the fast-atrophy class. CONCLUSIONS Heterogeneity of atrophic rates at the MCI stage is associated with several plasma proteins related to microglia activity, which show either protective or adverse effects on long-term cognition depending on the variability of atrophic rates.
Collapse
Affiliation(s)
- Jingyi Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Zhiyu Cao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Qun Yu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yingren Mai
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China
| | - Wang Liao
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Yuting Ruan
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen City, Guangdong Province, MN 518000, China; Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, MN 999077, China
| | - Lianhong Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou City, Guangdong Province, MN 510120, China.
| | - Jun Liu
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, No.250 East Changgang Road, Guangzhou City, Guangdong Province, MN 510260, China.
| |
Collapse
|
6
|
Lee JY, Lim MCX, Koh RY, Tsen MT, Chye SM. Blood-based therapies to combat neurodegenerative diseases. Metab Brain Dis 2024; 39:985-1004. [PMID: 38842660 DOI: 10.1007/s11011-024-01368-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Neurodegeneration, known as the progressive loss of neurons in terms of their structure and function, is the principal pathophysiological change found in the majority of brain-related disorders. Ageing has been considered the most well-established risk factor in most common neurodegenerative diseases, such as Parkinson's disease (PD) and Alzheimer's disease (AD). There is currently no effective treatment or cure for these diseases; the approved therapeutic options to date are only for palliative care. Ageing and neurodegenerative diseases are closely intertwined; reversing the aspects of brain ageing could theoretically mitigate age-related neurodegeneration. Ever since the regenerative properties of young blood on aged tissues came to light, substantial efforts have been focused on identifying and characterizing the circulating factors in the young and old systemic milieu that may attenuate or accentuate brain ageing and neurodegeneration. Later studies discovered the superiority of old plasma dilution in tissue rejuvenation, which is achieved through a molecular reset of the systemic proteome. These findings supported the use of therapeutic blood exchange for the treatment of degenerative diseases in older individuals. The first objective of this article is to explore the rejuvenating properties of blood-based therapies in the ageing brains and their therapeutic effects on AD. Then, we also look into the clinical applications, various limitations, and challenges associated with blood-based therapies for AD patients.
Collapse
Affiliation(s)
- Jia Yee Lee
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Mervyn Chen Xi Lim
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Min Tze Tsen
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
7
|
Yu Y, Yu S, Battaglia G, Tian X. Amyloid-β in Alzheimer's disease: Structure, toxicity, distribution, treatment, and prospects. IBRAIN 2024; 10:266-289. [PMID: 39346788 PMCID: PMC11427815 DOI: 10.1002/ibra.12155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/01/2024]
Abstract
Amyloid-β (Aβ) is a pivotal biomarker in Alzheimer's disease (AD), attracting considerable attention from numerous researchers. There is uncertainty regarding whether clearing Aβ is beneficial or harmful to cognitive function. This question has been a central topic of research, especially given the lack of success in developing Aβ-targeted drugs for AD. However, with the Food and Drug Administration's approval of Lecanemab as the first anti-Aβ medication in July 2023, there is a significant shift in perspective on the potential of Aβ as a therapeutic target for AD. In light of this advancement, this review aims to illustrate and consolidate the molecular structural attributes and pathological ramifications of Aβ. Furthermore, it elucidates the determinants influencing its expression levels while delineating the gamut of extant Aβ-targeted pharmacotherapies that have been subjected to clinical or preclinical evaluation. Subsequently, a comprehensive analysis is presented, dissecting the research landscape of Aβ across the domains above, culminating in the presentation of informed perspectives. Concluding reflections contemplate the supplementary advantages conferred by nanoparticle constructs, conceptualized within the framework of multivalent theory, within the milieu of AD diagnosis and therapeutic intervention, supplementing conventional modalities.
Collapse
Affiliation(s)
- Yifan Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shilong Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Giuseppe Battaglia
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Xiaohe Tian
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
8
|
Kumari P, Panigrahi AR, Yadav P, Beura SK, Singh SK. Platelets and inter-cellular communication in immune responses: Dialogue with both professional and non-professional immune cells. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:347-379. [PMID: 38762274 DOI: 10.1016/bs.apcsb.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Platelets, derived from bone marrow megakaryocytes, are essential for vascular integrity and play multifaceted roles in both physiological and pathological processes within the vasculature. Despite their small size and absence of a nucleus, platelets are increasingly recognized for their diverse immune functions. Recent research highlights their pivotal role in interactions with various immune cells, including professional cells like macrophages, dendritic cells, natural killer cells, T cells, and B cells, influencing host immune responses. Platelets also engage with non-professional immune cells, contributing to immune responses and structural maintenance, particularly in conditions like inflammation and atherosclerosis. This review underscores the emerging significance of platelets as potent immune cells, elucidating their interactions with the immune system. We explore the mechanisms of platelet activation, leading to diverse functions, such as aggregation, immunity, activation of other immune cells, and pathogen clearance. Platelets have become the predominant immune cells in circulation, involved in chronic inflammation, responses to infections, and autoimmune disorders. Their immunological attributes, including bioactive granule molecules and immune receptors, contribute to their role in immune responses. Unlike professional antigen-presenting cells, platelets process and present antigens through an MHC-I-dependent pathway, initiating T-cell immune responses. This review illuminates the unique features of platelets and their central role in modulating host immune responses in health and disease.
Collapse
Affiliation(s)
- Puja Kumari
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | | | - Pooja Yadav
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Samir Kumar Beura
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India
| | - Sunil Kumar Singh
- Department of Zoology, Central University of Punjab, Bathinda, Punjab, India; Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
9
|
Kobayashi M, Moro N, Yoshino A, Kumagawa T, Shijo K, Maeda T, Oshima H. Inhibition of P2X4 and P2X7 receptors improves histological and behavioral outcomes after experimental traumatic brain injury in rats. Exp Ther Med 2023; 26:378. [PMID: 37456165 PMCID: PMC10347371 DOI: 10.3892/etm.2023.12077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Release of large amounts of adenosine triphosphate (ATP), a gliotransmitter, into the extracellular space by traumatic brain injury (TBI) is considered to activate the microglia followed by release of inflammatory cytokines resulting in excessive inflammatory response that induces secondary brain injury. The present study investigated whether antagonists of ATP receptors (P2X4 and/or P2X7) on microglia are beneficial for reducing the post-injury inflammatory response that leads to secondary injury, a prognostic aggravation factor of TBI. Adult male Sprague-Dawley rats were subjected to cortical contusion injury (CCI) and randomly assigned to injury and drug treatment conditions, as follows: i) No surgical intervention (naïve group); ii) dimethyl sulfoxide treatment after CCI (CCI-control group); iii) 5-BDBD (antagonist of P2X4 receptor) treatment after CCI (CCI-5-BDBD group); iv) CCI-AZ11645373 (antagonist of P2X7 receptor) treatment after CCI (CCI-AZ11645373 group); v) or 5-BDBD and AZ11645373 treatment after CCI (CCI-5-BDBD + AZ11645373 group). In the CCI-5-BDBD, CCI-AZ11645373, and CCI-5-BDBD + AZ11645373 groups, expression of activated microglia was suppressed in the ipsilateral cortex and hippocampus 3 days after the CCI. Western blotting with ionized calcium-binding adaptor molecule 1 antibody revealed that administration of CCI-5-BDBD and/or CCI-AZ11645373 suppressed expression of microglia and reduced expression of inflammatory cytokine mRNA 3 days after the CCI. Furthermore, the plus maze test, which reflects the spatial memory function and involves the hippocampal function, showed improvement 28 days after secondary injury to the hippocampus. These findings confirmed that blocking the P2X4 and P2X7 receptors, which are ATP receptors central in gliotransmission, suppresses microglial activation and subsequent cytokine expression after brain injury, and demonstrates the potential as an effective treatment for reducing secondary brain injury.
Collapse
Affiliation(s)
- Masato Kobayashi
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Nobuhiro Moro
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
- Department of Neurological Surgery, Honjo-General Hospital, Saitama 367-0031, Japan
| | - Atsuo Yoshino
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takahiro Kumagawa
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Katsunori Shijo
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takeshi Maeda
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hideki Oshima
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
10
|
Jung MJ, Kim N, Jeon SH, Gee MS, Kim JW, Lee JK. Eugenol relieves the pathological manifestations of Alzheimer's disease in 5×FAD mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154930. [PMID: 37348246 DOI: 10.1016/j.phymed.2023.154930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/27/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the accumulation of amyloid-β (Aβ) and excessive neuroinflammation, resulting in neuronal cell death and cognitive impairments. Eugenol, a phenylpropene, is the main component of Syzygium aromaticum L. (Myrtaceae) and has multiple therapeutic effects, including neuroprotective and anti-inflammatory effects, through multimodal mechanisms. PURPOSE We aimed to investigate the effect of eugenol on AD pathologies using a 5× familiar AD (5×FAD) mouse model. METHODS Eight-month-old 5×FAD and wild-type mice were administered with eugenol (10 or 30 mg/kg/day, p.o) for 2 months. Y-maze and Morris water maze tests were performed to assess the cognitive function of mice. After the behavioral test, molecular analysis was conducted to investigate the therapeutic mechanism of eugenol. RESULTS Our findings indicate that eugenol treatment effectively mitigated cognitive impairments in 5×FAD mice. This beneficial effect was associated with a decrease in AD pathologies, including neuronal cell loss and Aβ deposition. Specifically, eugenol inhibited necroptosis activation and increased microglial phagocytosis, which were the underlying mechanisms for the observed reductions in neuronal cell loss and Aβ deposition, respectively. CONCLUSION Overall, our data suggest that eugenol would be a potential therapeutic candidate for AD.
Collapse
Affiliation(s)
- Min-Ji Jung
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Namkwon Kim
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Seung Ho Jeon
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Min Sung Gee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ji-Woon Kim
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea.
| | - Jong Kil Lee
- Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Huang Z. A Function of Amyloid-β in Mediating Activity-Dependent Axon/Synapse Competition May Unify Its Roles in Brain Physiology and Pathology. J Alzheimers Dis 2023; 92:29-57. [PMID: 36710681 PMCID: PMC10023438 DOI: 10.3233/jad-221042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Amyloid-β protein precursor (AβPP) gives rise to amyloid-β (Aβ), a peptide at the center of Alzheimer's disease (AD). AβPP, however, is also an ancient molecule dating back in evolution to some of the earliest forms of metazoans. This suggests a possible ancestral function that may have been obscured by those that evolve later. Based on literature from the functions of Aβ/AβPP in nervous system development, plasticity, and disease, to those of anti-microbial peptides (AMPs) in bacterial competition as well as mechanisms of cell competition uncovered first by Drosophila genetics, I propose that Aβ/AβPP may be part of an ancient mechanism employed in cell competition, which is subsequently co-opted during evolution for the regulation of activity-dependent neural circuit development and plasticity. This hypothesis is supported by foremost the high similarities of Aβ to AMPs, both of which possess unique, opposite (i.e., trophic versus toxic) activities as monomers and oligomers. A large body of data further suggests that the different Aβ oligomeric isoforms may serve as the protective and punishment signals long predicted to mediate activity-dependent axonal/synaptic competition in the developing nervous system and that the imbalance in their opposite regulation of innate immune and glial cells in the brain may ultimately underpin AD pathogenesis. This hypothesis can not only explain the diverse roles observed of Aβ and AβPP family molecules, but also provide a conceptual framework that can unify current hypotheses on AD. Furthermore, it may explain major clinical observations not accounted for and identify approaches for overcoming shortfalls in AD animal modeling.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
12
|
Suzuki C, Yamaguchi J, Sanada T, Oliva Trejo JA, Kakuta S, Shibata M, Tanida I, Uchiyama Y. Lack of Cathepsin D in the central nervous system results in microglia and astrocyte activation and the accumulation of proteinopathy-related proteins. Sci Rep 2022; 12:11662. [PMID: 35804072 PMCID: PMC9270453 DOI: 10.1038/s41598-022-15805-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/29/2022] [Indexed: 11/09/2022] Open
Abstract
Neuronal ceroid lipofuscinosis is one of many neurodegenerative storage diseases characterized by excessive accumulation of lipofuscins. CLN10 disease, an early infantile neuronal ceroid lipofuscinosis, is associated with a gene that encodes cathepsin D (CtsD), one of the major lysosomal proteases. Whole body CtsD-knockout mice show neurodegenerative phenotypes with the accumulation of lipofuscins in the brain and also show defects in other tissues including intestinal necrosis. To clarify the precise role of CtsD in the central nervous system (CNS), we generated a CNS-specific CtsD-knockout mouse (CtsD-CKO). CtsD-CKO mice were born normally but developed seizures and their growth stunted at around postnatal day 23 ± 1. CtsD-CKO did not exhibit apparent intestinal symptoms as those observed in whole body knockout. Histologically, autofluorescent materials were detected in several areas of the CtsD-CKO mouse's brain, including: thalamus, cerebral cortex, hippocampus, and cerebellum. Expression of ubiquitin and autophagy-associated proteins was also increased, suggesting that the autophagy-lysosome system was impaired. Microglia and astrocytes were activated in the CtsD-CKO thalamus, and inducible nitric oxide synthase (iNOS), an inflammation marker, was increased in the microglia. Interestingly, deposits of proteinopathy-related proteins, phosphorylated α-synuclein, and Tau protein were also increased in the thalamus of CtsD-CKO infant mice. Considering these results, we propose thatt the CtsD-CKO mouse is a useful mouse model to investigate the contribution of cathepsin D to the early phases of neurodegenerative diseases in relation to lipofuscins, proteinopathy-related proteins and activation of microglia and astrocytes.
Collapse
Affiliation(s)
- Chigure Suzuki
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Junji Yamaguchi
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Laboratory of Morphology and Image Analysis, Biomedical Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takahito Sanada
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Juan Alejandro Oliva Trejo
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Souichirou Kakuta
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.,Laboratory of Morphology and Image Analysis, Biomedical Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Masahiro Shibata
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima-shi, Kagoshima, 890-8544, Japan
| | - Isei Tanida
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
13
|
Situ M, Citalan-Madrid AF, Stamatovic SM, Keep RF, Andjelkovic AV. Transcriptomic Profile of Blood–Brain Barrier Remodeling in Cerebral Amyloid Angiopathy. Front Cell Neurosci 2022; 16:931247. [PMID: 35813502 PMCID: PMC9257207 DOI: 10.3389/fncel.2022.931247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/30/2022] [Indexed: 12/16/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a small vessel disease characterized by amyloid β (Aβ) peptide deposition within the walls of medium to small-caliber blood vessels, cerebral microhemorrhage, and blood–brain barrier (BBB) leakage. It is commonly associated with late-stage Alzheimer’s disease. BBB dysfunction is indicated as a pathological substrate for CAA progression with hyperpermeability, enhancing the extravasation of plasma components and inducing neuroinflammation, further worsening BBB injury and contributing to cognitive decline. Although significant effort has been made in defining the gene mutations and risk factors involved in microvascular alterations with vascular dementia and Alzheimer’s disease, the intra- and intercellular pathogenic mechanisms responsible for vascular hyperpermeability are still largely unknown. The present study aimed to elucidate the transcriptional profile of the cerebral microvessels (BBB) in a murine model with CAA vasculopathy to define potential causes and underlying mechanisms of BBB injury. A comprehensive RNA sequencing analysis was performed of CAA vasculopathy in Tg-SwDI mice at 6 and 18 months in comparison to age-matched wildtype controls to examine how age and amyloid accumulation impact the transcriptional signature of the BBB. Results indicate that Aβ has a critical role in triggering brain endothelial cell and BBB dysfunction in CAA vasculopathy, causing an intense proinflammatory response, impairing oxidative metabolism, altering the coagulation status of brain endothelial cells, and remodeling barrier properties. The proinflammatory response includes both adaptive and innate immunity, with pronounced induction of genes that regulate macrophage/microglial activation and chemokines/adhesion molecules that support T and B cell transmigration. Age has an important impact on the effects of Aβ, increasing the BBB injury in CAA vasculopathy. However, early inflammation, particularly microglia/macrophage activation and the mediators of B lymphocytes’ activities are underlying processes of BBB hyperpermeability and cerebral microbleeds in the early stage of CAA vasculopathy. These findings reveal a specific profile of the CAA-associated BBB injury that leads to a full progression of CAA.
Collapse
Affiliation(s)
- Muyu Situ
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- *Correspondence: Anuska V. Andjelkovic,
| |
Collapse
|
14
|
Rajesh Y, Kanneganti TD. Innate Immune Cell Death in Neuroinflammation and Alzheimer's Disease. Cells 2022; 11:1885. [PMID: 35741014 PMCID: PMC9221514 DOI: 10.3390/cells11121885] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder molecularly characterized by the formation of amyloid β (Aβ) plaques and type 2 microtubule-associated protein (Tau) abnormalities. Multiple studies have shown that many of the brain's immunological cells, specifically microglia and astrocytes, are involved in AD pathogenesis. Cells of the innate immune system play an essential role in eliminating pathogens but also regulate brain homeostasis and AD. When activated, innate immune cells can cause programmed cell death through multiple pathways, including pyroptosis, apoptosis, necroptosis, and PANoptosis. The cell death often results in the release of proinflammatory cytokines that propagate the innate immune response and can eliminate Aβ plaques and aggregated Tau proteins. However, chronic neuroinflammation, which can result from cell death, has been linked to neurodegenerative diseases and can worsen AD. Therefore, the innate immune response must be tightly balanced to appropriately clear these AD-related structural abnormalities without inducing chronic neuroinflammation. In this review, we discuss neuroinflammation, innate immune responses, inflammatory cell death pathways, and cytokine secretion as they relate to AD. Therapeutic strategies targeting these innate immune cell death mechanisms will be critical to consider for future preventive or palliative treatments for AD.
Collapse
|
15
|
Fakih W, Zeitoun R, AlZaim I, Eid AH, Kobeissy F, Abd-Elrahman KS, El-Yazbi AF. Early metabolic impairment as a contributor to neurodegenerative disease: Mechanisms and potential pharmacological intervention. Obesity (Silver Spring) 2022; 30:982-993. [PMID: 35470973 DOI: 10.1002/oby.23400] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 12/18/2022]
Abstract
The metabolic syndrome comprises a family of clinical and laboratory findings, including insulin resistance, hyperglycemia, hypertriglyceridemia, low high-density lipoprotein cholesterol levels, and hypertension, in addition to central obesity. The syndrome confers a high risk of cardiovascular mortality. Indeed, metabolic dysfunction has been shown to cause a direct insult to smooth muscle and endothelial components of the vasculature, which leads to vascular dysfunction and hyperreactivity. This, in turn, causes cerebral vasoconstriction and hypoperfusion, eventually contributing to cognitive deficits. Moreover, the metabolic syndrome disrupts key homeostatic processes in the brain, including apoptosis, autophagy, and neurogenesis. Impairment of such processes in the context of metabolic dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer, Parkinson, and Huntington diseases. The aim of this review is to elucidate the role that the metabolic syndrome plays in the pathogenesis of the latter disorders, with a focus on the role of perivascular adipose inflammation in the peripheral-to-central transduction of the inflammatory insult. This review delineates common signaling pathways that contribute to these pathologies. Moreover, the role of therapeutic agents aimed at treating the metabolic syndrome, as well as their risk factors that interfere with the aforementioned pathways, are discussed as potential interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Walaa Fakih
- Faculty of Pharmacy, Federation of Translational Medicine of Strasbourg, University of Strasbourg, Illkirch, France
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ralph Zeitoun
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, Psychiatry, Neuroscience and Chemistry, University of Florida, Gainesville, Florida, USA
| | - Khaled S Abd-Elrahman
- Brain and Mind Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmacology and Therapeutics, College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alamein International University, New Alamein City, Egypt
| |
Collapse
|
16
|
Wang Q, Yao H, Liu W, Ya B, Cheng H, Xing Z, Wu Y. Microglia Polarization in Alzheimer's Disease: Mechanisms and a Potential Therapeutic Target. Front Aging Neurosci 2021; 13:772717. [PMID: 34819850 PMCID: PMC8606412 DOI: 10.3389/fnagi.2021.772717] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/07/2021] [Indexed: 11/13/2022] Open
Abstract
Neuroinflammation regulated by microglia is one of the important factors involved in the pathogenesis of Alzheimer’s disease (AD). Activated microglia exhibited phenotypes termed as M1 and M2 phenotypes separately. M1 microglia contribute to the development of inflammation via upregulating pro-inflammatory cytokines, while M2 microglia exert anti-inflammation effects through enhancing the expression of anti-inflammation factors. Moreover, M1 and M2 microglia could be mutually transformed under various conditions. Both M1 and M2 microglia are implicated in AD. Amyloid-β (Aβ) and hyperphosphorylated tau are two major components of AD pathological hallmarks, neuritic plaques, and neurofibrillary tangles. Both Aβ and hyperphosphorylated tau were involved in microglial activation and subsequent inflammation, which further contribute to neuronal and synaptic loss in AD. In this review, we summarized the roles of M1 and M2 microglia in AD and underlying mechanisms, which will provide an insight into the role of microglia in the pathogenesis of AD and highlight the therapeutic potential of modulating microglia.
Collapse
Affiliation(s)
- Qinqin Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Hongmei Yao
- Affiliated Hospital of Jining Medical University, Jining, China
| | - Wenyan Liu
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Bailiu Ya
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Hongju Cheng
- Department of Physiology, College of Basic Medicine, Jining Medical University, Jining, China
| | - Zhenkai Xing
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- The Affiliated Kangning Hospital, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, School of Mental Health, Wenzhou Medical University, Wenzhou, China.,Oujiang Laboratory, Wenzhou, China
| |
Collapse
|
17
|
Busse S, Hoffmann J, Michler E, Hartig R, Frodl T, Busse M. Dementia-associated changes of immune cell composition within the cerebrospinal fluid. Brain Behav Immun Health 2021; 14:100218. [PMID: 34589754 PMCID: PMC8474581 DOI: 10.1016/j.bbih.2021.100218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 11/17/2022] Open
Abstract
Inflammation and alterations in essential protein structures in the brain might also change the cellular distribution in the cerebrospinal fluid (CSF). Using flow cytometry, we analyzed cell populations of the innate and adaptive immune system associated with the most frequent forms of dementias. We included patients with mild cognitive impairment (MCI; N = 33), Alzheimer’s disease (AD; N = 90), vascular dementia (VD; N = 35) and frontotemporal dementia (FTD; N = 17) at the time of diagnosis, before onset of treatment and 11 elderly non-demented individuals. Dependent on the form of dementia, an increased frequency of CD14+ monocytes, NK cells and NKT cells was measured. Within the T cell population, a dementia-associated shift from central memory towards (late-stage) effector cells was detected. T cells and NKT cells were correlated with MMSE, NK and NKT cells were correlated with ptau, CD14+ monocytes and NK cells were correlated with Amyloid-β 1–40. Our data suggest that each investigated immune cell type is involved in dementia-associated alterations within the CSF, possibly having distinct functions in their pathogenesis.
Collapse
Affiliation(s)
- Stefan Busse
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Jessica Hoffmann
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Enrico Michler
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Roland Hartig
- Institute of Immunology, University of Magdeburg, Magdeburg, Germany
| | - Thomas Frodl
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Mandy Busse
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Corresponding author. Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, D-39120, Magdeburg, Germany.
| |
Collapse
|
18
|
Pototskiy E, Vinokuroff K, Ojeda A, Major CK, Sharma D, Anderson T, Howard K, Borenstein R, Musto AE. Downregulation of CD40L-CD40 attenuates seizure susceptibility and severity of seizures. Sci Rep 2021; 11:17262. [PMID: 34446808 PMCID: PMC8390750 DOI: 10.1038/s41598-021-96760-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/11/2021] [Indexed: 12/04/2022] Open
Abstract
Unregulated neuro-inflammation mediates seizures in temporal lobe epilepsy (TLE). Our aim was to determine the effect of CD40-CD40L activation in experimental seizures. CD40 deficient mice (CD40KO) and control mice (wild type, WT) received pentenyltetrazole (PTZ) or pilocarpine to evaluate seizures and status epilepticus (SE) respectively. In mice, anti-CD40L antibody was administered intranasally before PTZ. Brain samples from human TLE and post-seizure mice were processed to determine CD40-CD40L expression using histological and molecular techniques. CD40 expression was higher in hippocampus from human TLE and in cortical neurons and hippocampal neural terminals after experimental seizures. CD40-CD40L levels increased after seizures in the hippocampus and in the cortex. After SE, CD40L/CD40 levels increased in cortex and showed an upward trend in the hippocampus. CD40KO mice demonstrated reduction in seizure severity and in latency compared to WT mice. Anti-CD40L antibody limited seizure susceptibility and seizure severity. CD40L-CD40 interaction can serve as a target for an immuno-therapy for TLE.
Collapse
Affiliation(s)
- Esther Pototskiy
- Department of Pathology and Anatomy, Eastern Virginia Medical School, 700 W. Olney Road, Lewis Hall, Office 2174, Norfolk, VA, 23507, USA
| | | | - Andrew Ojeda
- Department of Pathology and Anatomy, Eastern Virginia Medical School, 700 W. Olney Road, Lewis Hall, Office 2174, Norfolk, VA, 23507, USA
| | | | | | | | | | - Ronen Borenstein
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Alberto E Musto
- Department of Pathology and Anatomy, Eastern Virginia Medical School, 700 W. Olney Road, Lewis Hall, Office 2174, Norfolk, VA, 23507, USA.
- Department of Neurology, Eastern Virginia Medical School, 700 W. Olney Road, Lewis Hall, Office 2174, Norfolk, VA, 23507, USA.
| |
Collapse
|
19
|
Vasilopoulou F, Escolano C, Pallàs M, Griñán-Ferré C. Microarray Analysis Revealed Inflammatory Transcriptomic Changes after LSL60101 Treatment in 5XFAD Mice Model. Genes (Basel) 2021; 12:1315. [PMID: 34573297 PMCID: PMC8468036 DOI: 10.3390/genes12091315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/23/2022] Open
Abstract
I2-IR have been found dysregulated in patients with neurodegenerative diseases, such as Alzheimer's disease (AD), in which the importance of neuroinflammation in the establishment and maintenance of cognitive decline is well-documented. To research the implication of I2-IR in neuroinflammatory pathways altered in AD, we determined the expression profile of genes associated with inflammation in the 5XFAD model treated with LSL60101, a well-established I2-IR ligand. Thus, we performed a qPCR array containing 84 inflammation-related genes. Hierarchical clustering analysis revealed three gene clusters, suggesting that treatment with LSL60101 affects the gene expression associated with inflammation in the 5XFAD model. Furthermore, we evaluated the functions of the three clusters; thereby performing a pathway enrichment analysis using the GO database. As we expected, clusters 2 and 3 showed alterations in the inflammatory response, chemotaxis and the chemokine-mediated signaling pathway, among others. To validate previous results from the gene profiling analysis, the expression levels of a representative subset of mRNAs were selected according to the intensity of the observed changes and their biological relevance. Interestingly, changes induced by LSL60101 in the 5XFAD model were validated for several genes. These results suggest that treatment with LSL60101 in the 5XFAD model reverses the inflammatory process during the development of AD.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain;
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, University of Barcelona (NeuroUB), Av. Joan XXIII 27-31, 08028 Barcelona, Spain; (F.V.); (M.P.)
| |
Collapse
|
20
|
Yasuno F, Nakamura A, Kato T, Iwata K, Sakurai T, Arahata Y, Washimi Y, Hattori H, Ito K. An evaluation of the amyloid cascade model using in vivo positron emission tomographic imaging. Psychogeriatrics 2021; 21:14-23. [PMID: 32783314 DOI: 10.1111/psyg.12589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/03/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022]
Abstract
AIM The amyloid cascade hypothesis posits that the accumulation of amyloid β (Aβ) is the triggering factor for Alzheimer's disease, which consecutively induces aggregation of tau, synaptic loss, and cell death. Most experimental and clinical evidence supports this model, but the available data are largely qualitative. Here, we tested the amyloid cascade hypothesis by using in vivo evaluation of positron emission tomography and magnetic resonance imaging. METHODS Path analysis was used to estimate the relationships among Aβ accumulation (PiB standardized uptake value ratio (SUVR)), tau aggregation and its related neuroinflammation (THK5351 SUVR), grey matter atrophy in the medial temporal region, and memory function in Aβ-positive subjects. We also performed additional regression analyses to evaluate the effect of Aβ on the toxicity of tau aggregation/neuroinflammation. RESULTS Path analysis supported our hypothesized model: Aβ accumulation affected tau aggregation/neuroinflammation in the medial temporal region, and these pathological changes caused of the grey matter atrophy and memory dysfunction. In separate regression analyses, THK5351 SUVR had a significant effect on grey matter atrophy only in PiB-positive subjects. The analysis of the interaction effect showed that the effects of THK5351 SUVR on grey matter atrophy were significantly different between PiB-positive and PiB-negative groups. When we included the effect of being an apolipoprotein E ε4 carrier as a covariate, the interaction effect remained significant. CONCLUSION Our in vivo evaluation of positron emission tomographic and magnetic resonance imaging data supported the amyloid cascade hypothesis. In addition, it indicated that Aβ not only accelerates tau aggregation/neuroinflammation but promotes its toxicity. Our findings showed the importance of understanding the role and therapeutic potential of the interaction between amyloid and tau aggregation/neuroinflammation in Alzheimer's disease.
Collapse
Affiliation(s)
- Fumihiko Yasuno
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan.,National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Akinori Nakamura
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan.,National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kaori Iwata
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Sakurai
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yutaka Arahata
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yukihiko Washimi
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Hideyuki Hattori
- National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Kengo Ito
- Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan.,National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, Obu, Japan
| |
Collapse
|
21
|
Rawish E, Nording H, Münte T, Langer HF. Platelets as Mediators of Neuroinflammation and Thrombosis. Front Immunol 2020; 11:548631. [PMID: 33123127 PMCID: PMC7572851 DOI: 10.3389/fimmu.2020.548631] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Beyond platelets function in hemostasis, there is emerging evidence to suggest that platelets contribute crucially to inflammation and immune responses. Therefore, considering the detrimental role of inflammatory conditions in severe neurological disorders such as multiple sclerosis or stroke, this review outlines platelets involvement in neuroinflammation. For this, distinct mechanisms of platelet-mediated thrombosis and inflammation are portrayed, focusing on the interaction of platelet receptors with other immune cells as well as brain endothelial cells. Furthermore, we draw attention to the intimate interplay between platelets and the complement system as well as between platelets and plasmatic coagulation factors in the course of neuroinflammation. Following the thorough exposition of preclinical approaches which aim at ameliorating disease severity after inducing experimental autoimmune encephalomyelitis (a counterpart of multiple sclerosis in mice) or brain ischemia-reperfusion injury, the clinical relevance of platelet-mediated neuroinflammation is addressed. Thus, current as well as future propitious translational and clinical strategies for the treatment of neuro-inflammatory diseases by affecting platelet function are illustrated, emphasizing that targeting platelet-mediated neuroinflammation could become an efficient adjunct therapy to mitigate disease severity of multiple sclerosis or stroke associated brain injury.
Collapse
Affiliation(s)
- Elias Rawish
- University Hospital Schleswig-Holstein, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Henry Nording
- University Hospital Schleswig-Holstein, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Thomas Münte
- University Hospital Schleswig-Holstein, Clinic for Neurology, Lübeck, Germany
| | - Harald F. Langer
- University Hospital Schleswig-Holstein, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
22
|
Vigil FA, Bozdemir E, Bugay V, Chun SH, Hobbs M, Sanchez I, Hastings SD, Veraza RJ, Holstein DM, Sprague SM, M Carver C, Cavazos JE, Brenner R, Lechleiter JD, Shapiro MS. Prevention of brain damage after traumatic brain injury by pharmacological enhancement of KCNQ (Kv7, "M-type") K + currents in neurons. J Cereb Blood Flow Metab 2020; 40:1256-1273. [PMID: 31272312 PMCID: PMC7238379 DOI: 10.1177/0271678x19857818] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nearly three million people in the USA suffer traumatic brain injury (TBI) yearly; however, there are no pre- or post-TBI treatment options available. KCNQ2-5 voltage-gated K+ channels underlie the neuronal "M current", which plays a dominant role in the regulation of neuronal excitability. Our strategy towards prevention of TBI-induced brain damage is predicated on the suggested hyper-excitability of neurons induced by TBIs, and the decrease in neuronal excitation upon pharmacological augmentation of M/KCNQ K+ currents. Seizures are very common after a TBI, making further seizures and development of epilepsy disease more likely. Our hypothesis is that TBI-induced hyperexcitability and ischemia/hypoxia lead to metabolic stress, cell death and a maladaptive inflammatory response that causes further downstream morbidity. Using the mouse controlled closed-cortical impact blunt TBI model, we found that systemic administration of the prototype M-channel "opener", retigabine (RTG), 30 min after TBI, reduces the post-TBI cascade of events, including spontaneous seizures, enhanced susceptibility to chemo-convulsants, metabolic stress, inflammatory responses, blood-brain barrier breakdown, and cell death. This work suggests that acutely reducing neuronal excitability and energy demand via M-current enhancement may be a novel model of therapeutic intervention against post-TBI brain damage and dysfunction.
Collapse
Affiliation(s)
- Fabio A Vigil
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Eda Bozdemir
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Vladislav Bugay
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Sang H Chun
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - MaryAnn Hobbs
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Isamar Sanchez
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shayne D Hastings
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Rafael J Veraza
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Deborah M Holstein
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shane M Sprague
- Department of Neurosurgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Chase M Carver
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jose E Cavazos
- Department of Neurology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Robert Brenner
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - James D Lechleiter
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mark S Shapiro
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
23
|
Pereira CF, Santos AE, Moreira PI, Pereira AC, Sousa FJ, Cardoso SM, Cruz MT. Is Alzheimer's disease an inflammasomopathy? Ageing Res Rev 2019; 56:100966. [PMID: 31577960 DOI: 10.1016/j.arr.2019.100966] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/28/2019] [Accepted: 09/27/2019] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly and, despite the tremendous efforts researchers have put into AD research, there are no effective options for prevention and treatment of the disease. The best way to reach this goal is to clarify the mechanisms involved in the onset and progression of AD. In the last few years the views about the drivers of AD have been changing and nowadays it is believed that neuroinflammation takes center stage in disease pathogenesis. Herein, we provide an overview about the role of neuroinflammation in AD describing the role of microglia and astroglia is this process. Then, we will debate the NLRP3 inflammasome putting the focus on its activation through the canonical, non-canonical and alternative pathways and the triggers involved herein namely endoplasmic reticulum stress, mitochondrial dysfunction, reactive oxygen species and amyloid β peptide. Data supporting the hypothesis that inflammasome-mediated peripheral inflammation may contribute to AD pathology will be presented. Finally, a brief discussion about the therapeutic potential of NLRP3 inflammasome modulation is also provided.
Collapse
|
24
|
Cassano T, Calcagnini S, Carbone A, Bukke VN, Orkisz S, Villani R, Romano A, Avolio C, Gaetani S. Pharmacological Treatment of Depression in Alzheimer's Disease: A Challenging Task. Front Pharmacol 2019; 10:1067. [PMID: 31611786 PMCID: PMC6777507 DOI: 10.3389/fphar.2019.01067] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/21/2019] [Indexed: 12/17/2022] Open
Abstract
Besides the memory impairment, Alzheimer’s disease (AD) is often complicated by neuropsychiatric symptoms also known as behavioral and psychological symptoms of dementia, which occur in one-third of patients at an early stage of the disease. Although the relationship between depressive disorders and AD is debated, the question if depression is a prodromal symptom preceding cognitive deficits or an independent risk factor for AD is still unclear. Moreover, there is growing evidence reporting that conventional antidepressants are not effective in depression associated with AD and, therefore, there is an urgent need to understand the neurobiological mechanism underlying the resistance to the antidepressants. Another important question that remains to be addressed is whether the antidepressant treatment is able to modulate the levels of amyloid-β peptide (Aβ), which is a key pathological hallmark in AD. The present review summarizes the present knowledge on the link between depression and AD with a focus on the resistance of antidepressant therapies in AD patients. Finally, we have briefly outlined the preclinical and clinical evidences behind the possible mechanisms by which antidepressants modulate Aβ pathology. To our opinion, understanding the cellular processes that regulate Aβ levels may provide greater insight into the disease pathogenesis and might be helpful in designing novel selective and effective therapy against depression in AD.
Collapse
Affiliation(s)
- Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Silvio Calcagnini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Antonio Carbone
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Vidyasagar Naik Bukke
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stanislaw Orkisz
- Morphological Science Department of Human Anatomy, Medical Faculty, University of Rzeszów, Rzeszów, Poland
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Carlo Avolio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Meyer PF, Savard M, Poirier J, Morgan D, Breitner J. Hypothesis: cerebrospinal fluid protein markers suggest a pathway toward symptomatic resilience to AD pathology. Alzheimers Dement 2019; 15:1160-1171. [PMID: 31405825 DOI: 10.1016/j.jalz.2019.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION We sought biological pathways that explained discordance between Alzheimer's disease (AD) pathology and symptoms. METHODS In 306 Alzheimer's Disease Neuroimaging Initiative (ADNI)-1 participants across the AD clinical spectrum, we investigated association between cognitive outcomes and 23 cerebrospinal fluid (CSF) analytes associated with abnormalities in the AD biomarkers amyloid β1-42 and total-tau. In a 200-person "training" set, Least Absolute Shrinkage and Selection Operator regression estimated model weights for the 23 proteins, and for the AD biomarkers themselves, as predictors of ADAS-Cog11 scores. In the remaining 106 participants ("validation" set), fully adjusted regression models then tested the Least Absolute Shrinkage and Selection Operator-derived models and a related protein marker summary score as predictors of ADAS-Cog11, ADNI diagnostic category, and longitudinal cognitive trajectory. RESULTS AD biomarkers alone explained 26% of the variance in validation set cognitive scores. Surprisingly, the 23 AD-related proteins explained 31% of this variance. The biomarkers and protein markers appeared independent in this respect, jointly explaining 42% of test score variance. The composite protein marker score also predicted ADNI diagnosis and subsequent cognitive trajectory. Cognitive outcome prediction redounded principally to ten markers related to lipid or vascular functions or to microglial activation or chemotaxis. In each analysis, apoE protein and four markers in the latter immune-activation group portended better outcomes. DISCUSSION CSF markers of vascular, lipid-metabolic and immune-related functions may explain much of the disjunction between AD biomarker abnormality and symptom severity. In particular, our results suggest the hypothesis that innate immune activation improves cognitive outcomes in persons with AD pathology. This hypothesis should be tested by further study of cognitive outcomes related to CSF markers of innate immune activation.
Collapse
Affiliation(s)
- Pierre-François Meyer
- Faculty of Medicine, McGill University, Montréal, QC, Canada; Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Melissa Savard
- McGill Center for Studies on Aging, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Judes Poirier
- Faculty of Medicine, McGill University, Montréal, QC, Canada; Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada; Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada
| | - David Morgan
- College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Faculty of Medicine, McGill University, Montréal, QC, Canada; Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada; Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada.
| | | |
Collapse
|
26
|
Gao G, Zhao S, Xia X, Li C, Li C, Ji C, Sheng S, Tang Y, Zhu J, Wang Y, Huang Y, Zheng JC. Glutaminase C Regulates Microglial Activation and Pro-inflammatory Exosome Release: Relevance to the Pathogenesis of Alzheimer's Disease. Front Cell Neurosci 2019; 13:264. [PMID: 31316350 PMCID: PMC6611423 DOI: 10.3389/fncel.2019.00264] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/28/2019] [Indexed: 01/02/2023] Open
Abstract
Microglial activation is a key pathogenic process at the onset of Alzheimer’s disease (AD). Identifying regulators of microglial activation bears great potential in elucidating causes and mechanisms of AD and determining candidates for early intervention. Previous studies demonstrate abnormal elevation of glutaminase C (GAC) in HIV-infected or immune-activated microglia. However, whether GAC elevation causes microglial activation remains unknown. In this study, we found heightened expression levels of GAC in early AD mouse brain tissues compared with those in control littermates. Investigations on an in vitro neuroinflammation model revealed that GAC is increased in primary mouse microglia following pro-inflammatory stimulation. To model GAC elevation we overexpressed GAC by plasmid transfection and observed that GAC-overexpression shift the microglial phenotype to a pro-inflammatory state. Treatment with BPTES, a glutaminase inhibitor, reversed LPS-induced microglial activation and inflammation. Furthermore, we discovered that GAC overexpression in mouse microglia increased exosome release and changed exosome content, which includes specific packaging of pro-inflammatory miRNAs that activate microglia. Together, our results demonstrate a causal effect of GAC elevation on microglial activation and exosome release, both of which promote the establishment of a pro-inflammatory microenvironment. Therefore, GAC may have important relevance to the pathogenesis of AD.
Collapse
Affiliation(s)
- Ge Gao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Shu Zhao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chunhong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Congcong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chenhui Ji
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shiyang Sheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yalin Tang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jie Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States.,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Meyer PF, Savard M, Poirier J, Labonté A, Rosa-Neto P, Weitz TM, Town T, Breitner J. Bi-directional Association of Cerebrospinal Fluid Immune Markers with Stage of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2019; 63:577-590. [PMID: 29660934 PMCID: PMC5929310 DOI: 10.3233/jad-170887] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immune mechanisms may be important in the pathogenesis of Alzheimer’s disease (AD). Yet, studies comparing cerebrospinal fluid (CSF) and plasma immune marker levels of healthy and demented individuals have yielded conflicting results. We analyzed CSF from 101 members of the parental history-positive PREVENT-AD cohort of healthy aging adults, and 237 participants without dementia from the initial cohort of the Alzheimer’s Disease Neuroimaging Initiative (ADNI-1). Following recent practice, we used the biomarkers total-tau and amyloid-β1-42 to allocate participants from each study into four stages of AD pathogenesis: Stage 0 (no abnormality), Stage 1 (reduced amyloid-β1-42), Stage 2 (reduced amyloid-β1-42 and increased total-tau), or “Suspected Non-Alzheimer Pathology” (elevated total-tau only). Investigating the PREVENT-AD participants’ CSF assay results for 19 immune/inflammatory markers, we found six that showed a distinct bi-directional relationship with pathogenetic stage. Relative to Stage 0, these were diminished at Stage 1 but strongly increased at Stage 2. Among the ADNI participants (90 healthy controls and 147 with mild cognitive impairment), we found that 23 of 83 available CSF markers also showed this distinct pattern. These results support recent observations that immune activation may become apparent only after the onset of both amyloid and tau pathologies. Unexpectedly, they also suggest that immune marker activity may diminish along with earliest appearance of amyloid-β plaque pathology. These findings may explain discordant results from past studies, and suggest the importance of characterizing the extent of AD pathology when comparing clinical groups.
Collapse
Affiliation(s)
- Pierre-François Meyer
- Faculty of Medicine, McGill University, Montréal, QC, Canada.,Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Melissa Savard
- Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada.,McGill Centre for Studies in Aging and Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Judes Poirier
- Faculty of Medicine, McGill University, Montréal, QC, Canada.,Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada.,Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada
| | - Anne Labonté
- Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada.,Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada
| | - Pedro Rosa-Neto
- Faculty of Medicine, McGill University, Montréal, QC, Canada.,Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada.,Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada.,McGill Centre for Studies in Aging and Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Tara M Weitz
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Terrence Town
- Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - John Breitner
- Faculty of Medicine, McGill University, Montréal, QC, Canada.,Center for Studies on the Prevention of Alzheimer's Disease (StoP-AD), Douglas Mental Health University Institute, Montréal, QC, Canada.,Douglas Mental Health University Institute Research Centre, Montréal, QC, Canada.,McGill Centre for Studies in Aging and Douglas Mental Health University Institute, Montréal, QC, Canada
| | | | | |
Collapse
|
28
|
El Hajj H, Savage JC, Bisht K, Parent M, Vallières L, Rivest S, Tremblay MÈ. Ultrastructural evidence of microglial heterogeneity in Alzheimer's disease amyloid pathology. J Neuroinflammation 2019; 16:87. [PMID: 30992040 PMCID: PMC6469225 DOI: 10.1186/s12974-019-1473-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease, characterized by the deposition of extracellular fibrillar amyloid β (fΑβ) and the intracellular accumulation of neurofibrillary tangles. As AD progresses, Aβ drives a robust and prolonged inflammatory response via its recognition by microglia, the brain's immune cells. Microglial reactivity to fAβ plaques may impair their normal surveillance duties, facilitating synaptic loss and neuronal death, as well as cognitive decline in AD. METHODS In the current study, we performed correlative light, transmission, and scanning electron microscopy to provide insights into microglial structural and functional heterogeneity. We analyzed microglial cell bodies and processes in areas containing fAβ plaques and neuronal dystrophy, dystrophy only, or appearing healthy, among the hippocampus CA1 of 14-month-old APPSwe-PS1Δe9 mice versus wild-type littermates. RESULTS Our quantitative analysis revealed that microglial cell bodies in the AD model mice were larger and displayed ultrastructural signs of cellular stress, especially nearby plaques. Microglial cell bodies and processes were overall less phagocytic in AD model mice. However, they contained increased fibrillar materials and non-empty inclusions proximal to plaques. Microglial cell bodies and processes in AD model mice also displayed reduced association with extracellular space pockets that contained debris. In addition, microglial processes in healthy subregions of AD model mice encircled synaptic elements more often compared with plaque-associated processes. These observations in mice were qualitatively replicated in post-mortem hippocampal samples from two patients with AD (Braak stage 5). CONCLUSION Together, our findings identify at the ultrastructural level distinct microglial transformations common to mouse and human in association with amyloid pathology.
Collapse
Affiliation(s)
- Hassan El Hajj
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Julie C. Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Kanchan Bisht
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Martin Parent
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Quebec, QC Canada
- CERVO Brain Research Center, Quebec, QC Canada
| | - Luc Vallières
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| | - Serge Rivest
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| |
Collapse
|
29
|
Gyengesi E, Rangel A, Ullah F, Liang H, Niedermayer G, Asgarov R, Venigalla M, Gunawardena D, Karl T, Münch G. Chronic Microglial Activation in the GFAP-IL6 Mouse Contributes to Age-Dependent Cerebellar Volume Loss and Impairment in Motor Function. Front Neurosci 2019; 13:303. [PMID: 31001075 PMCID: PMC6456818 DOI: 10.3389/fnins.2019.00303] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/18/2019] [Indexed: 11/15/2022] Open
Abstract
Chronic microglial activation is a prominent feature of many chronic neurodegenerative diseases, including Parkinson’s and Alzheimer’s disease. To investigate the effects of chronic microglial activation on cerebellar structure and motor function throughout the lifespan, the transgenic GFAP-IL6 mouse model was used. The aim of the study was to examine inflammatory markers and neuronal degeneration while simultaneously characterizing the motor performance of GFAP-IL6 mice at 3, 6, 14, and 24 months of age in comparison to WT (C57BL/6) mice. In respect to markers of neuroinflammation in the cerebellum, increased numbers of Iba1+ microglia were observed as early as at 3 months of age. In addition, TNF-α levels proved to be significantly higher in the GFAP-IL6 compared to WT mice at all time points. A difference in cerebellar volume between the GFAP-IL6 and WT mice was observed later in life, starting at 6 months and increasing to a loss of about 50% in aged (24 months old) GFAP-IL6 mice. Synaptic deficits were also assessed by using pre- (synaptophysin) and post-synaptic (PSD95) markers. While synaptophysin levels remained unchanged, PSD95 levels decreased in the aging GFAP-IL6 mice compared to their WT littermates from 14 months onward. To assess the effect of microglia activation and neurodegeneration on behavior, a variety of motor function tests, semi-quantitative cerebellar ataxia score, accelerod, beam walking, and open field tests were performed. An age-dependent difference between the genotypes was observed in many of the motor function tests. For example, reduced performance on the accelerod and higher ataxia scores were observed at 6 months of age, followed by the beam walking test showing differences at 14 months of age. In summary, this study constitutes a comprehensive, age-dependent examination of inflammatory, synaptic and neurodegenerative changes in the brains of GFAP-IL6 mice leading to a deterioration in motor performance. The results also indicate that early chronic microglia activation in the GFAP-IL6 mouse leads to observable cerebellar volume loss and motor deficits later in life.
Collapse
Affiliation(s)
- Erika Gyengesi
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Alejandra Rangel
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Faheem Ullah
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Huazheng Liang
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,Department of Neurology, Shanghai Fourth People's Hospital, Tongji University, Shanghai, China
| | - Garry Niedermayer
- School of Science and Health, Western Sydney University, Penrith, NSW, Australia
| | - Rustam Asgarov
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Madhuri Venigalla
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Dhanushka Gunawardena
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Tim Karl
- Behavioral Neuroscience Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
30
|
Adlimoghaddam A, Snow WM, Stortz G, Perez C, Djordjevic J, Goertzen AL, Ko JH, Albensi BC. Regional hypometabolism in the 3xTg mouse model of Alzheimer's disease. Neurobiol Dis 2019; 127:264-277. [PMID: 30878533 DOI: 10.1016/j.nbd.2019.03.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 02/22/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive age-related neurodegenerative disease. Although neurofibrillary tangles and amyloid beta are classic hallmarks of AD, the earliest deficits in AD progression may be caused by unknown factors. One suspected factor has to do with brain energy metabolism. To investigate this factor, brain metabolic activity in 3xTg-AD mice and age-matched controls were measured with FDG-PET. Significant hypometabolic changes (p < .01) in brain metabolism were detected in the cortical piriform and insular regions of AD brains relative to controls. These regions are associated with olfaction, which is a potential clinical marker for AD progression as well as neurogenesis. The activity of the terminal component of the mitochondrial respiratory chain (complex IV) and the expression of complex I-V were significantly decreased (p < .05), suggesting that impaired metabolic activity coupled with impaired oxidative phosphorylation leads to decreased mitochondrial bioenergetics and subsequent Neurodegeneration. Although there is an association between neuroinflammatory pathological markers (microglial) and hypometabolism in AD, there was no association found between neuropathological (Aβ, tau, and astrocytes) and functional changes in AD sensitive brain regions, also suggesting that brain hypometabolism occurs prior to AD pathology. Therefore, targeting metabolic mechanisms in cortical piriform and insular regions at early stages may be a promising approach for preventing, slowing, and/or blocking the onset of AD and preserving neurogenesis.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada.
| | | | | | - Claudia Perez
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada
| | - Jelena Djordjevic
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada
| | | | - Ji Hyun Ko
- Dept. of Human Anatomy and Cell Science, University of Manitoba, Canada; Health Sciences Centre, Canada
| | - Benedict C Albensi
- St. Boniface Hospital Research, Canada; Dept. of Pharmacology & Therapeutics, University of Manitoba, Canada.
| |
Collapse
|
31
|
Huang Q, Sun D, Zubair Hussain M, Liu Y, A. Morozova-Roche L, Zhang C. HEWL interacts with dissipated oleic acid micelles, and decreases oleic acid cytotoxicity. PLoS One 2019; 14:e0212648. [PMID: 30794655 PMCID: PMC6386356 DOI: 10.1371/journal.pone.0212648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/06/2019] [Indexed: 11/18/2022] Open
Abstract
Senile plaques are well-known hallmarks of Alzheimer's Diseases (AD). However, drugs targeting tangles of the protein tau and plaques of β-amyloid have no significant effect on disease progression, and the studies on the underlying mechanism of AD remain in high demand. Growing evidence supports the protective role of senile plaques in local inflammation driven by S100A9. We herein demonstrate that oleic acid (OA) micelles interact with hen egg white lysozyme (HEWL) and promote its amyloid formation. Consequently, SH-SY5Y cell line and mouse neural stem cells are rescued from OA toxicity by co-aggregation of OA and HEWL. Using atomic force microscopy in combination with fluorescence microscopy, we revealed that HEWL forms round-shaped aggregates in the presence of OA micelles instead of protofibrils of HEWL alone. These HEWL amyloids act as a sink for toxic OA micelles and their co-aggregate form large clumps, suggesting a protective function in amyloid and OA cytotoxicity.
Collapse
Affiliation(s)
- Qin Huang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional Materials, Institute of Photonics and Photon-Technology, Northwest University, Xi’an, China
| | - Dan Sun
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional Materials, Institute of Photonics and Photon-Technology, Northwest University, Xi’an, China
| | - Muhammad Zubair Hussain
- Department of Zoology, Government Emerson College, Multan, Pakistan
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Yonggang Liu
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | | | - Ce Zhang
- State Key Laboratory of Cultivation Base for Photoelectric Technology and Functional Materials, Institute of Photonics and Photon-Technology, Northwest University, Xi’an, China
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| |
Collapse
|
32
|
Co-signaling Molecules in Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:233-265. [PMID: 31758537 DOI: 10.1007/978-981-32-9717-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Cabinio M, Saresella M, Piancone F, LaRosa F, Marventano I, Guerini FR, Nemni R, Baglio F, Clerici M. Association between Hippocampal Shape, Neuroinflammation, and Cognitive Decline in Alzheimer’s Disease. J Alzheimers Dis 2018; 66:1131-1144. [DOI: 10.3233/jad-180250] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Monia Cabinio
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Marina Saresella
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Federica Piancone
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Francesca LaRosa
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Ivana Marventano
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Franca Rosa Guerini
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Raffaello Nemni
- Neurorehabilitation Unit, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Francesca Baglio
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Mario Clerici
- Laboratory of Molecular Medicine and Imaging in Rehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Ye X, Zhou W, Zhang J. Association of CSF CD40 levels and synaptic degeneration across the Alzheimer's disease spectrum. Neurosci Lett 2018; 694:41-45. [PMID: 30447377 DOI: 10.1016/j.neulet.2018.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/01/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023]
Abstract
The CD40 pathway has been implicated in microglial activation, which is considered as a key factor in the pathogenesis of Alzheimer's disease (AD). However, the association of CSF CD40 and synaptic degeneration in living human is not clear. A total of 294 subjects with different severities of cognitive impairments were included in this study: 84 participants with normal cognition, 143 patients with mild cognitive impairment (MCI) and 67 patients with mild AD. Levels of CD40 in CSF were compared among the three groups. Further, several linear regression models were conducted to explore the associations of CSF CD40 and neurogranin levels (reflecting synaptic degeneration) when controlling for age, gender, educational attainment, APOE4 genotype, clinical diagnosis, CSF Aβ42 and tau proteins. We found that CSF CD40 levels were significantly decreased in patients with mild AD compared with healthy controls and MCI patients (control vs. AD, p = 0.0026; MCI vs. AD, p = 0.0268). However, there were no significant differences in CSF CD40 levels between controls and patients with MCI (p = 0.37). In addition, CSF CD40 levels were associated with neurogranin in the pooled sample when controlling for age, gender, educational attainment, APOE4 genotype and diagnosis. In summary, our findings support the notion that the CD40 pathway may contribute to an important mechanism underlying synaptic degeneration in AD.
Collapse
Affiliation(s)
- Xinwu Ye
- Wenzhou Seventh People's Hospital, Zhejiang, China.
| | - Wenjun Zhou
- Department of Pathology, Hangzhou Normal University, College of Medicine, Zhejiang, China
| | - Jie Zhang
- Independent Researcher, 25 Xuezheng road, Xiasha District, Zhejiang, 310018, China.
| |
Collapse
|
35
|
Mustaly-Kalimi S, Littlefield AM, Stutzmann GE. Calcium Signaling Deficits in Glia and Autophagic Pathways Contributing to Neurodegenerative Disease. Antioxid Redox Signal 2018; 29:1158-1175. [PMID: 29634342 DOI: 10.1089/ars.2017.7266] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Numerous cellular processes and signaling mechanisms have been identified that contribute to Alzheimer's disease (AD) pathology; however, a comprehensive or unifying pathway that binds together the major disease features remains elusive. As an upstream mechanism, altered calcium (Ca2+) signaling is a common driving force for many pathophysiological events that emerge during normal aging and development of neurodegenerative disease. Recent Advances: Over the previous three decades, accumulated evidence has validated the concept that intracellular Ca2+ dysregulation is centrally involved in AD pathogenesis, including the aggregation of pathogenic β-amyloid (Aβ) and phospho-τ species, synapse loss and dysfunction, cognitive impairment, and neurotoxicity. CRITICAL ISSUES Although neuronal Ca2+ signaling within the cytosol and endoplasmic reticulum (ER) has been well studied, other critical central nervous system-resident cell types affected by aberrant Ca2+ signaling, such as astrocytes and microglia, have not been considered as thoroughly. In addition, certain intracellular Ca2+-harboring organelles have been well studied, such as the ER and mitochondria; however other critical Ca2+-regulated organelles, such as lysosomes and autophagosomes, have only more recently been investigated. In this review, we examine Ca2+ dysregulation in microglia and astrocytes, as well as key intracellular organelles important for cellular maintenance and protein handling. Ca2+ dysregulation within these non-neuronal cells and organelles is hypothesized to disrupt the effective clearance of misaggregated proteins and cellular signaling pathways needed for memory networks. FUTURE DIRECTIONS Overall, we aim to explore how these disrupted mechanisms could be involved in AD pathology and consider their role as potential therapeutic targets. Antioxid. Redox Signal. 29, 1158-1175.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Alyssa M Littlefield
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Grace E Stutzmann
- 2 Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| |
Collapse
|
36
|
Busse M, Michler E, von Hoff F, Dobrowolny H, Hartig R, Frodl T, Busse S. Alterations in the Peripheral Immune System in Dementia. J Alzheimers Dis 2018; 58:1303-1313. [PMID: 28582858 DOI: 10.3233/jad-161304] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Alterations in the immune response that result in inflammation might play a role in the pathology of dementias. In order to analyze changes of the peripheral immune system associated with different types of dementias, we determined several innate and adaptive cell populations in whole blood using flow cytometry. We included patients with Alzheimer's disease (AD; n = 60), vascular dementia (VaD; n = 20), and frontotemporal dementia (FTD; n = 12) at the time point of diagnosis and 24 age-matched neuropsychiatric healthy persons. Monocytes and NK cells were diminished in VaD, but not in AD and FTD. B cell and T cell numbers were decreased in all investigated forms of dementia. Changes in the contribution of naïve/memory T cells were only present in AD. Correlation and regression analyses revealed associations between altered immune cell populations and Q Albumin as marker for the integrity of the blood-cerebrospinal fluid-barrier, Mini-Mental State Examination values, and age. The peripheral immune system is altered in AD, VaD, and FTD. However, each disorder presents unique changes in the investigated cell types indicating different mechanisms underlying the pathology.
Collapse
Affiliation(s)
- Mandy Busse
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Enrico Michler
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Franz von Hoff
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Roland Hartig
- Institute of Immunology, University of Magdeburg, Magdeburg, Germany
| | - Thomas Frodl
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| | - Stefan Busse
- Department of Psychiatry, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
37
|
Protective effects of evodiamine in experimental paradigm of Alzheimer's disease. Cogn Neurodyn 2018; 12:303-313. [PMID: 29765479 DOI: 10.1007/s11571-017-9471-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 10/23/2017] [Accepted: 12/29/2017] [Indexed: 01/07/2023] Open
Abstract
Evodiamine, a major component of Evodia rutaecarpa, has been reported to possess various pharmacological activities, including anti-inflammatory, antioxidative stress, and neuroprotective effects. Our previous study has shown that the potential effects of evodiamine on the learning and memory impairments in the transgenic mouse model of Alzheimer's disease (AD). The present study was designed to investigate neuroprotective mechanism and therapeutic potential of evodiamine against intracerebroventricular streptozotocin (ICV-STZ)-induced experimental sporadic Alzheimer's disease in mice. STZ was injected twice intracerebroventrically (3 mg/kg ICV) on alternate days (day 1 and day 3) in mice. Daily oral administration with evodiamine (50 or 100 mg/kg per day) starting from the first dose of STZ for 21 days showed an improvement in STZ induced cognitive deficits as assessed by novel object recognition and Morris water maze test. Evodiamine significantly decreased STZ induced elevation in acetylcholinesterase activity and malondialdehyde level, and significantly increased STZ induced reduction in glutathione activities and superoxide dismutase activities in the hippocampus compared to control. Furthermore, evodiamine inhibited significantly glial cell activation and neuroinflammation (TNF-α, IL-1β, and IL-6 levels) in the hippocampus. Moreover, evodiamine increased the activity of AKT/GSK-3β signalling pathway and inhibited the activity of nuclear factor κB. In summary, our study suggests that evodiamine can be a novel therapeutic agent for the management of sporadic AD.
Collapse
|
38
|
Pandey A, Bani S, Dutt P, Kumar Satti N, Avtar Suri K, Nabi Qazi G. Multifunctional neuroprotective effect of Withanone, a compound from Withania somnifera roots in alleviating cognitive dysfunction. Cytokine 2017; 102:211-221. [PMID: 29108796 DOI: 10.1016/j.cyto.2017.10.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 09/28/2017] [Accepted: 10/21/2017] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is a chronic disorder that slowly worsens and impairs the person's memory, learning, reasoning, judgment, communication and familiar tasks with loss of orientation. AD is characterized clinically by cognitive deficit and pathologically by the deposition of β amyloid plaques, neurofibrillary tangles, associated with degeneration of the cholinergic forebrain. Withanone (WS-2), a compound isolated from root extract of Withania somnifera at doses administered orally/day to wistar rats for duration of 21 days showed significant improvement in the cognitive skill by inhibiting amyloid β-42 and attenuated the elevated levels of pro-inflammatory cytokines like TNF alpha, IL-1 beta, IL-6, MCP-1, Nitric oxide, lipid peroxidation and both β- and γ- secretase enzymatic activity. Administration of WS-2 also significantly reversed the decline in acetyl choline and Glutathione (GSH) activity. None of the treatments that are available today alter the underlying causes of this terminal disease. Few preliminary clinical treatments have demonstrated that some plant medicines do ameliorate and improve memory and learning in patients with mild-to-moderate AD. WS-2 showed promise in AD treatment because of cognitive benefits and more importantly, mechanisms of action with respect to the fundamental pathophysiology of the disease, not limited to the inhibition of AChE, but also include the modification of Aβ processing, protection against oxidative stress and anti-inflammatory effects.
Collapse
Affiliation(s)
- Anjali Pandey
- Pharmacology Divison, Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, Jammu and Kashmir State, India.
| | - Sarang Bani
- Pharmacology Divison, Indian Institute of Integrative Medicine, Canal Road, Jammu-180001, Jammu and Kashmir State, India.
| | - Prabhu Dutt
- Natural product chemistry divison, Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi-180001, Jammu and Kashmir State, India.
| | - Naresh Kumar Satti
- Natural product chemistry divison, Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi-180001, Jammu and Kashmir State, India.
| | - Krishan Avtar Suri
- Natural product chemistry divison, Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi-180001, Jammu and Kashmir State, India.
| | - Ghulam Nabi Qazi
- Natural product chemistry divison, Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi-180001, Jammu and Kashmir State, India.
| |
Collapse
|
39
|
M344 promotes nonamyloidogenic amyloid precursor protein processing while normalizing Alzheimer's disease genes and improving memory. Proc Natl Acad Sci U S A 2017; 114:E9135-E9144. [PMID: 29073110 PMCID: PMC5664514 DOI: 10.1073/pnas.1707544114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Hundreds of failed clinical trials with Alzheimer’s disease (AD) patients over the last fifteen years demonstrate that the one-target–one-disease approach is not effective in AD. In silico, structure-based, multitarget drug design approaches to treat multifactorial diseases have not been successful in the context of AD either. Here, we show that M344, an inhibitor of class I and IIB histone deacetylases, affects multiple AD-related genes, including those related to both early- and late-onset AD. We also show that M344 improves memory in the 3xTg AD mouse model. This work endorses a shift to a multitargeted approach to the treatment of AD, supporting the therapeutic potential of a single small molecule with an epigenetic mechanism of action. Alzheimer’s disease (AD) comprises multifactorial ailments for which current therapeutic strategies remain insufficient to broadly address the underlying pathophysiology. Epigenetic gene regulation relies upon multifactorial processes that regulate multiple gene and protein pathways, including those involved in AD. We therefore took an epigenetic approach where a single drug would simultaneously affect the expression of a number of defined AD-related targets. We show that the small-molecule histone deacetylase inhibitor M344 reduces beta-amyloid (Aβ), reduces tau Ser396 phosphorylation, and decreases both β-secretase (BACE) and APOEε4 gene expression. M344 increases the expression of AD-relevant genes: BDNF, α-secretase (ADAM10), MINT2, FE65, REST, SIRT1, BIN1, and ABCA7, among others. M344 increases sAPPα and CTFα APP metabolite production, both cleavage products of ADAM10, concordant with increased ADAM10 gene expression. M344 also increases levels of immature APP, supporting an effect on APP trafficking, concurrent with the observed increase in MINT2 and FE65, both shown to increase immature APP in the early secretory pathway. Chronic i.p. treatment of the triple transgenic (APPsw/PS1M146V/TauP301L) mice with M344, at doses as low as 3 mg/kg, significantly prevented cognitive decline evaluated by Y-maze spontaneous alternation, novel object recognition, and Barnes maze spatial memory tests. M344 displays short brain exposure, indicating that brief pulses of daily drug treatment may be sufficient for long-term efficacy. Together, these data show that M344 normalizes several disparate pathogenic pathways related to AD. M344 therefore serves as an example of how a multitargeting compound could be used to address the polygenic nature of multifactorial diseases.
Collapse
|
40
|
Takkinen JS, López-Picón FR, Al Majidi R, Eskola O, Krzyczmonik A, Keller T, Löyttyniemi E, Solin O, Rinne JO, Haaparanta-Solin M. Brain energy metabolism and neuroinflammation in ageing APP/PS1-21 mice using longitudinal 18F-FDG and 18F-DPA-714 PET imaging. J Cereb Blood Flow Metab 2017; 37:2870-2882. [PMID: 27834284 PMCID: PMC5536795 DOI: 10.1177/0271678x16677990] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preclinical animal model studies of brain energy metabolism and neuroinflammation in Alzheimer's disease have produced conflicting results, hampering both the elucidation of the underlying disease mechanism and the development of effective Alzheimer's disease therapies. Here, we aimed to quantify the relationship between brain energy metabolism and neuroinflammation in the APP/PS1-21 transgenic mouse model of Alzheimer's disease using longitudinal in vivo18F-FDG and 18F-DPA-714) PET imaging and ex vivo brain autoradiography. APP/PS1-21 (TG, n = 9) and wild type control mice (WT, n = 9) were studied longitudinally every third month from age 6 to 15 months with 18F-FDG and 18F-DPA-714 with a one-week interval between the scans. Additional TG (n = 52) and WT (n = 29) mice were used for ex vivo studies. In vivo, the 18F-FDG SUVs were lower and the 18F-DPA-714 binding ratios relative to the cerebellum were higher in the TG mouse cortex and hippocampus than in WT mice at age 12 to 15 months ( p < 0.05). The ex vivo cerebellum binding ratios supported the results of the in vivo18F-DPA-714 studies but not the 18F-FDG studies. This longitudinal PET study demonstrated decreased energy metabolism and increased inflammation in the brains of APP/PS1-21 mice compared to WT mice.
Collapse
Affiliation(s)
- Jatta S Takkinen
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Francisco R López-Picón
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Rana Al Majidi
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| | - Olli Eskola
- 2 Turku PET Centre, University of Turku, Turku, Finland
| | | | - Thomas Keller
- 2 Turku PET Centre, University of Turku, Turku, Finland
| | | | - Olof Solin
- 2 Turku PET Centre, University of Turku, Turku, Finland.,4 Turku PET Centre, Åbo Akademi University, Turku, Finland.,5 Department of Chemistry, University of Turku, Turku, Finland
| | - Juha O Rinne
- 6 Turku PET Centre, Turku University Hospital, Turku, Finland.,7 Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- 1 MediCity Research Laboratory, University of Turku, Turku, Finland.,2 Turku PET Centre, University of Turku, Turku, Finland
| |
Collapse
|
41
|
Habib A, Sawmiller D, Li S, Xiang Y, Rongo D, Tian J, Hou H, Zeng J, Smith A, Fan S, Giunta B, Mori T, Currier G, Shytle DR, Tan J. LISPRO mitigates β-amyloid and associated pathologies in Alzheimer's mice. Cell Death Dis 2017; 8:e2880. [PMID: 28617434 PMCID: PMC5520933 DOI: 10.1038/cddis.2017.279] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/28/2017] [Accepted: 05/12/2017] [Indexed: 01/09/2023]
Abstract
Lithium has been marketed in the United States of America since the 1970s as a treatment for bipolar disorder. More recently, studies have shown that lithium can improve cognitive decline associated with Alzheimer’s disease (AD). However, the current United States Food and Drug Administration-approved lithium pharmaceutics (carbonate and citrate chemical forms) have a narrow therapeutic window and unstable pharmacokinetics that, without careful monitoring, can cause serious adverse effects. Here, we investigated the safety profile, pharmacokinetics, and therapeutic efficacy of LISPRO (ionic co-crystal of lithium salicylate and l-proline), lithium salicylate, and lithium carbonate (Li2CO3). We found that LISPRO (8-week oral treatment) reduces β-amyloid plaques and phosphorylation of tau by reducing neuroinflammation and inactivating glycogen synthase kinase 3β in transgenic Tg2576 mice. Specifically, cytokine profiles from the brain, plasma, and splenocytes suggested that 8-week oral treatment with LISPRO downregulates pro-inflammatory cytokines, upregulates anti-inflammatory cytokines, and suppresses renal cyclooxygenase 2 expression in transgenic Tg2576 mice. Pharmacokinetic studies indicated that LISPRO provides significantly higher brain lithium levels and more steady plasma lithium levels in both B6129SF2/J (2-week oral treatment) and transgenic Tg2576 (8-week oral treatment) mice compared with Li2CO3. Oral administration of LISPRO for 28 weeks significantly reduced β-amyloid plaques and tau-phosphorylation. In addition, LISPRO significantly elevated pre-synaptic (synaptophysin) and post-synaptic protein (post synaptic density protein 95) expression in brains from transgenic 3XTg-AD mice. Taken together, our data suggest that LISPRO may be a superior form of lithium with improved safety and efficacy as a potential new disease modifying drug for AD.
Collapse
Affiliation(s)
- Ahsan Habib
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Darrell Sawmiller
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Song Li
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Yang Xiang
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - David Rongo
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jun Tian
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Huayan Hou
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jin Zeng
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adam Smith
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Shengnuo Fan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Brian Giunta
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Takashi Mori
- Departments of Biomedical Sciences and Pathology, Saitama Medical Center and Saitama Medical University, Kawagoe, Saitama, Japan
| | - Glenn Currier
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Douglas Ronald Shytle
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Jun Tan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center, Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
42
|
Gaikwad S, Patel D, Agrawal-Rajput R. CD40 Negatively Regulates ATP-TLR4-Activated Inflammasome in Microglia. Cell Mol Neurobiol 2017; 37:351-359. [PMID: 26961545 PMCID: PMC11482137 DOI: 10.1007/s10571-016-0358-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 02/29/2016] [Indexed: 01/01/2023]
Abstract
During acute brain injury and/or sterile inflammation, release of danger-associated molecular patterns (DAMPs) activates pattern recognition receptors (PRRs). Microglial toll-like receptor (TLR)-4 activated by DAMPs potentiates neuroinflammation through inflammasome-induced IL-1β and pathogenic Th17 polarization which critically influences brain injury. TLR4 activation accompanies increased CD40, a cognate costimulatory molecule, involved in microglia-mediated immune responses in the brain. During brain injury, excessive release of extracellular ATP (DAMPs) is involved in promoting the damage. However, the regulatory role of CD40 in microglia during ATP-TLR4-mediated inflammasome activation has never been explored. We report that CD40, in the absence of ATP, synergizes TLR4-induced proinflammatory cytokines but not IL-1β, suggesting that the response is independent of inflammasome. The presence of ATP during TLR4 activation leads to NLRP3 inflammasome activation and caspase-1-mediated IL-1β secretion which was inhibited during CD40 activation, accompanied with inhibition of ERK1/2 and reactive oxygen species (ROS), and elevation in p38 MAPK phosphorylation. Experiments using selective inhibitors prove indispensability of ERK 1/2 and ROS for inflammasome activation. The ATP-TLR4-primed macrophages polarize the immune response toward pathogenic Th17 cells, whereas CD40 activation mediates Th1 response. Exogenous supplementation of IFN-γ (a Th1 cytokine and CD40 inducer) results in decreased IL-1β, suggesting possible feedback loop mechanism of inflammasome inhibition, whereby IFN-γ-mediated increase in CD40 expression and activation suppress neurotoxic inflammasome activation required for Th17 response. Collectively, the findings indicate that CD40 is a novel negative regulator of ATP-TLR4-mediated inflammasome activation in microglia, thus providing a checkpoint to regulate excessive inflammasome activation and Th17 response during DAMP-mediated brain injury.
Collapse
Affiliation(s)
- Sagar Gaikwad
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India
| | - Divyesh Patel
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India
| | - Reena Agrawal-Rajput
- Department of Immunology, School of Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382 007, India.
| |
Collapse
|
43
|
Bioactive Nutrients and Nutrigenomics in Age-Related Diseases. Molecules 2017; 22:molecules22010105. [PMID: 28075340 PMCID: PMC6155887 DOI: 10.3390/molecules22010105] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 12/20/2016] [Accepted: 01/03/2017] [Indexed: 01/10/2023] Open
Abstract
The increased life expectancy and the expansion of the elderly population are stimulating research into aging. Aging may be viewed as a multifactorial process that results from the interaction of genetic and environmental factors, which include lifestyle. Human molecular processes are influenced by physiological pathways as well as exogenous factors, which include the diet. Dietary components have substantive effects on metabolic health; for instance, bioactive molecules capable of selectively modulating specific metabolic pathways affect the development/progression of cardiovascular and neoplastic disease. As bioactive nutrients are increasingly identified, their clinical and molecular chemopreventive effects are being characterized and systematic analyses encompassing the "omics" technologies (transcriptomics, proteomics and metabolomics) are being conducted to explore their action. The evolving field of molecular pathological epidemiology has unique strength to investigate the effects of dietary and lifestyle exposure on clinical outcomes. The mounting body of knowledge regarding diet-related health status and disease risk is expected to lead in the near future to the development of improved diagnostic procedures and therapeutic strategies targeting processes relevant to nutrition. The state of the art of aging and nutrigenomics research and the molecular mechanisms underlying the beneficial effects of bioactive nutrients on the main aging-related disorders are reviewed herein.
Collapse
|
44
|
Shukla M, Govitrapong P, Boontem P, Reiter RJ, Satayavivad J. Mechanisms of Melatonin in Alleviating Alzheimer's Disease. Curr Neuropharmacol 2017; 15:1010-1031. [PMID: 28294066 PMCID: PMC5652010 DOI: 10.2174/1570159x15666170313123454] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 02/10/2017] [Accepted: 03/09/2017] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic, progressive and prevalent neurodegenerative disease characterized by the loss of higher cognitive functions and an associated loss of memory. The thus far "incurable" stigma for AD prevails because of variations in the success rates of different treatment protocols in animal and human studies. Among the classical hypotheses explaining AD pathogenesis, the amyloid hypothesis is currently being targeted for drug development. The underlying concept is to prevent the formation of these neurotoxic peptides which play a central role in AD pathology and trigger a multispectral cascade of neurodegenerative processes post-aggregation. This could possibly be achieved by pharmacological inhibition of β- or γ-secretase or stimulating the nonamyloidogenic α-secretase. Melatonin the pineal hormone is a multifunctioning indoleamine. Production of this amphiphilic molecule diminishes with advancing age and this decrease runs parallel with the progression of AD which itself explains the potential benefits of melatonin in line of development and devastating consequences of the disease progression. Our recent studies have revealed a novel mechanism by which melatonin stimulates the nonamyloidogenic processing and inhibits the amyloidogenic processing of β-amyloid precursor protein (βAPP) by stimulating α -secretases and consequently down regulating both β- and γ-secretases at the transcriptional level. In this review, we discuss and evaluate the neuroprotective functions of melatonin in AD pathogenesis, including its role in the classical hypotheses in cellular and animal models and clinical interventions in AD patients, and suggest that with early detection, melatonin treatment is qualified to be an anti-AD therapy.
Collapse
Affiliation(s)
- Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakornpathom 73170, Thailand
| | - Parichart Boontem
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 54 Kamphaeng Phet 6 Road, Lak Si, Bangkok10210, Thailand
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jutamaad Satayavivad
- Chulabhorn Research Institute and Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok10210, Thailand
| |
Collapse
|
45
|
Zhang W, Zhi D, Ren H, Wang D, Wang X, Zhang Z, Fei D, Zhu H, Li H. Shengmai Formula Ameliorates Pathological Characteristics in AD C. elegans. Cell Mol Neurobiol 2016; 36:1291-1302. [PMID: 26886750 PMCID: PMC11482344 DOI: 10.1007/s10571-015-0326-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/25/2015] [Indexed: 12/26/2022]
Abstract
Shengmai (SM) formula, a classical traditional Chinese medicine formula, is composed of Panax ginseng (Pg), Ophiopogon japonicus (Oj), and Schisandra Chinesis (Sc). SM has been clinically used to treat heart failure and ischemic heart disease. Although SM formula has been reported to be potential for fighting against Alzheimer's disease (AD) by previous works, there are many gaps in our knowledge on its usage in AD treatment on an organism level and will then need to be further clarified. In this study, transgenic Caenorhabditis elegans expressing human Aβ1-42 are used to evaluate SM formula efficacy to treat AD phenotype and to investigate its underlying mechanism. The results showed that SM formula ameliorated AD pathological characteristics of paralysis behavior and chemotaxis defect in transgenic C. elegans. With SM treatment, the number of Aβ deposits decreased, the levels of gene expressions of hsp16-2, hsp16-41, ace-1, ace-2, and TNFA1P1 homolog genes were down-regulated. Our results also showed that Oj exhibited more stronger effect on delaying paralysis in worms than Pg and Sc did, and synergistic action was observed between Pg and Oj, and Sc further enhanced the activity of Pg/Oj combination on delaying paralysis behavior. Further, SM with herbs of Pg, Oj, and Sc at a dose proportion of 9:9:6 exhibited superior therapeutic efficacy in comparison with herbs at other dose proportions. After SM formula extracted by ethanol, it delayed AD symptoms on a wider dose from 0.2 to 10.0 mg/mL with no toxic effect. These results provided more evidence for SM formula being potential to be used to treat AD.
Collapse
Affiliation(s)
- Weimin Zhang
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Dejuan Zhi
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Hui Ren
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Dong Wang
- Institute of Microbiology, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Xin Wang
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Zhanxin Zhang
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Dongqing Fei
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Hongmei Zhu
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China
| | - Hongyu Li
- Gansu High Throughput Screening and Creation Center for Health Products, School of Pharmacy, Lanzhou University, No. 199, Donggang West Road, Lanzhou, 730000, China.
- Institute of Microbiology, School of Life Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
46
|
Rajasekar N, Nath C, Hanif K, Shukla R. Intranasal Insulin Administration Ameliorates Streptozotocin (ICV)-Induced Insulin Receptor Dysfunction, Neuroinflammation, Amyloidogenesis, and Memory Impairment in Rats. Mol Neurobiol 2016; 54:6507-6522. [PMID: 27730514 DOI: 10.1007/s12035-016-0169-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is associated with reduced insulin level and impairment of insulin receptor (IR) signaling in the brain, which correlates to amyloid pathology, neuroinflammation, and synaptic neurotoxicity. Clinical studies show that intranasal insulin improves memory in AD patients without peripheral hypoglycemia. However, neuroprotective molecular mechanism of the beneficial effect of intranasal insulin in AD pathology is unexplored. Therefore, we investigated the role of intranasal insulin on intracerebroventricular (ICV) streptozotocin (STZ)-induced memory impairment in rats as evaluated in the Morris water maze test. STZ (ICV) treated rats had shown memory impairment along with a significant decrease in IR signaling molecules (IR, pIRS-1, pAkt, and pGSK-3α/β expression) and IDE expression in both hippocampus and cerebral cortex. Intranasal insulin delivery prevented these changes. Moreover, intranasal insulin was found to inhibit significantly glial cell activation (GFAP and Iba-1 expression), neuroinflammation (COX-2 expression, NFκB translocation, TNF-α, and IL-10 level) and amyloidogenic protein expression (BACE-1 and Aβ1-42 expression) in STZ (ICV)-injected rats. STZ (ICV)-induced caspase activation and postsynaptic neurotoxicity were also prevented by treatment with intranasal insulin. Our findings reveal that insulin has the neuroprotective effect and clearly signifies the potential use of intranasal insulin delivery for the treatment of AD. Graphical Abstract Neuroprotective effects of intranasal insulin administration on streptozotocin (ICV)-induced memory impairment in rats.
Collapse
Affiliation(s)
- N Rajasekar
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Chandishwar Nath
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Kashif Hanif
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India
| | - Rakesh Shukla
- Divisions of Pharmacology and Toxicology, CSIR-Central Drug Research Institute, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Chennai, India.
| |
Collapse
|
47
|
Badea A, Kane L, Anderson RJ, Qi Y, Foster M, Cofer GP, Medvitz N, Buckley AF, Badea AK, Wetsel WC, Colton CA. The fornix provides multiple biomarkers to characterize circuit disruption in a mouse model of Alzheimer's disease. Neuroimage 2016; 142:498-511. [PMID: 27521741 DOI: 10.1016/j.neuroimage.2016.08.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/23/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022] Open
Abstract
Multivariate biomarkers are needed for detecting Alzheimer's disease (AD), understanding its etiology, and quantifying the effect of therapies. Mouse models provide opportunities to study characteristics of AD in well-controlled environments that can help facilitate development of early interventions. The CVN-AD mouse model replicates multiple AD hallmark pathologies, and we identified multivariate biomarkers characterizing a brain circuit disruption predictive of cognitive decline. In vivo and ex vivo magnetic resonance imaging (MRI) revealed that CVN-AD mice replicate the hippocampal atrophy (6%), characteristic of humans with AD, and also present changes in subcortical areas. The largest effect was in the fornix (23% smaller), which connects the septum, hippocampus, and hypothalamus. In characterizing the fornix with diffusion tensor imaging, fractional anisotropy was most sensitive (20% reduction), followed by radial (15%) and axial diffusivity (2%), in detecting pathological changes. These findings were strengthened by optical microscopy and ultrastructural analyses. Ultrastructual analysis provided estimates of axonal density, diameters, and myelination-through the g-ratio, defined as the ratio between the axonal diameter, and the diameter of the axon plus the myelin sheath. The fornix had reduced axonal density (47% fewer), axonal degeneration (13% larger axons), and abnormal myelination (1.5% smaller g-ratios). CD68 staining showed that white matter pathology could be secondary to neuronal degeneration, or due to direct microglial attack. In conclusion, these findings strengthen the hypothesis that the fornix plays a role in AD, and can be used as a disease biomarker and as a target for therapy.
Collapse
Affiliation(s)
- Alexandra Badea
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA.
| | - Lauren Kane
- Trinity College of Arts & Sciences, Duke University, Durham, NC 27710, USA
| | - Robert J Anderson
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Yi Qi
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Mark Foster
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Gary P Cofer
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Neil Medvitz
- Department of Pathology, and Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Anne F Buckley
- Department of Pathology, and Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Andreas K Badea
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - William C Wetsel
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Carol A Colton
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
48
|
ElAli A, Rivest S. Microglia in Alzheimer's disease: A multifaceted relationship. Brain Behav Immun 2016; 55:138-150. [PMID: 26254232 DOI: 10.1016/j.bbi.2015.07.021] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting elderly people worldwide, which is mainly characterized by cerebral amyloid-beta (Aβ) plaque deposition and neurofibrillary tangle formation. The interest in microglia arose from the overwhelming experimental evidence that outlined a key role of neuroinflammation in AD pathology. Microglia constitute the powerhouse of the innate immune system in the brain. It is now widely accepted that microglia are myeloid-derived cells that infiltrate the developing brain at the early embryonic stages, and acquire a highly ramified phenotype postnatally. Microglia use these dynamic ramifications as sentinels to sense and detect any occurring alteration in brain homeostasis. Once a danger signal is detected, microglia get activated by acquiring a less ramified phenotype, and mount adequate responses that range from phagocyting cell debris to secreting inflammatory and trophic factors. Earlier reports have demonstrated, unequivocally, that microglia surround Aβ plaques and internalize Aβ microaggregates. However, the implication of these observations in AD pathology, and consequently treatment, is still a matter of debate. Nonetheless, targeting the activity of these cells constituted a convergent point in this debate. Unfortunately, the conflicting experimental findings obtained following the modulation of microglial activity in AD, further fueled the debate. This review aims at providing an overview regarding what we know about the implication of microglia in AD pathology, and treatment. The emerging role of monocytes is also discussed.
Collapse
Affiliation(s)
- Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Canada.
| |
Collapse
|
49
|
Chen W, Abud EA, Yeung ST, Lakatos A, Nassi T, Wang J, Blum D, Buée L, Poon WW, Blurton-Jones M. Increased tauopathy drives microglia-mediated clearance of beta-amyloid. Acta Neuropathol Commun 2016; 4:63. [PMID: 27339073 PMCID: PMC4918195 DOI: 10.1186/s40478-016-0336-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/13/2016] [Indexed: 12/23/2022] Open
Abstract
Alzheimer disease is characterized by the accumulation of β-amyloid (Aβ) plaques and tau-laden neurofibrillary tangles. Emerging studies suggest that in neurodegenerative diseases, aggregation of one protein species can promote other proteinopathies and that inflammation plays an important role in this process. To study the interplay between Aβ deposition, tau pathology, and microgliosis, we established a new AD transgenic mouse model by crossing 5xfAD mice with Thy-Tau22 transgenic mice. The resulting 'T5x' mice exhibit a greater than three-fold increase in misfolded and hyperphosphorylated tau and further substantiates the hypothesis that Aβ accelerates tau pathology. Surprisingly, T5x mice exhibit a 40-50 % reduction in Aβ plaque load and insoluble Aβ species when compared with aged-matched 5xfAD littermates. T5x mice exhibit significant changes in cytokine production, an almost doubling of microglial number, and a dramatic shift in microglia activation state. Furthermore, T5x microglia exhibit increased phagocytic capacity that enhances the clearance of insoluble Aβ and decreasing plaque load. Therefore, our results suggest that strategies to increase the phagocytic ability of microglia can be employed to reduce Aβ and that tau-induced changes in microglial activation state can promote the clearance of Aβ.
Collapse
|
50
|
Rani V, Deshmukh R, Jaswal P, Kumar P, Bariwal J. Alzheimer's disease: Is this a brain specific diabetic condition? Physiol Behav 2016; 164:259-67. [PMID: 27235734 DOI: 10.1016/j.physbeh.2016.05.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/16/2016] [Accepted: 05/22/2016] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2DM) are the two major health issues affecting millions of elderly people worldwide, with major impacts in the patient's daily life. Numerous studies have demonstrated that patients with diabetes have an increased risk of developing AD compared with healthy individuals. The principal biological mechanisms that associate with the progression of diabetes and AD are not completely understood. Impaired insulin signaling, uncontrolled glucose metabolism, oxidative stress, abnormal protein processing, and the stimulation of inflammatory pathways are common features to both AD and T2DM. In recent years brain specific abnormalities in insulin and insulin like growth factor (IGF) signaling considered as a major trigger involved in the etiopathogenesis of AD, showing T2DM like milieu. This review summarizes the pathways that might link diabetes and AD and the effect of diminished insulin.
Collapse
Affiliation(s)
- Vanita Rani
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Rahul Deshmukh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India.
| | - Priya Jaswal
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Puneet Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Jitender Bariwal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| |
Collapse
|