1
|
Pan Y, Li L, Cao N, Liao J, Chen H, Zhang M. Advanced nano delivery system for stem cell therapy for Alzheimer's disease. Biomaterials 2025; 314:122852. [PMID: 39357149 DOI: 10.1016/j.biomaterials.2024.122852] [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: 06/20/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's Disease (AD) represents one of the most significant neurodegenerative challenges of our time, with its increasing prevalence and the lack of curative treatments underscoring an urgent need for innovative therapeutic strategies. Stem cells (SCs) therapy emerges as a promising frontier, offering potential mechanisms for neuroregeneration, neuroprotection, and disease modification in AD. This article provides a comprehensive overview of the current landscape and future directions of stem cell therapy in AD treatment, addressing key aspects such as stem cell migration, differentiation, paracrine effects, and mitochondrial translocation. Despite the promising therapeutic mechanisms of SCs, translating these findings into clinical applications faces substantial hurdles, including production scalability, quality control, ethical concerns, immunogenicity, and regulatory challenges. Furthermore, we delve into emerging trends in stem cell modification and application, highlighting the roles of genetic engineering, biomaterials, and advanced delivery systems. Potential solutions to overcome translational barriers are discussed, emphasizing the importance of interdisciplinary collaboration, regulatory harmonization, and adaptive clinical trial designs. The article concludes with reflections on the future of stem cell therapy in AD, balancing optimism with a pragmatic recognition of the challenges ahead. As we navigate these complexities, the ultimate goal remains to translate stem cell research into safe, effective, and accessible treatments for AD, heralding a new era in the fight against this devastating disease.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| | - Long Li
- Department of Neurosurgery, First Hospital of China Medical University, Liaoning, 110001, China.
| | - Ning Cao
- Army Medical University, Chongqing, 400000, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Huiyue Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, 110001, China.
| | - Meng Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
2
|
Arnanz MA, Ferrer M, Grande MT, de Martín Esteban SR, Ruiz-Pérez G, Cravatt BF, Mostany R, Lobo VJSA, Romero J, Martínez-Relimpio AM. Fatty acid amide hydrolase gene inactivation induces hetero-cellular potentiation of microglial function in the 5xFAD mouse model of Alzheimer's disease. Glia 2025; 73:352-367. [PMID: 39474846 DOI: 10.1002/glia.24638] [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: 07/11/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 12/22/2024]
Abstract
Neuroinflammation has recently emerged as a crucial factor in Alzheimer's disease (AD) etiopathogenesis. Microglial cells play an important function in the inflammatory response; specifically, the emergence of disease-associated microglia (DAM) has offered new insights into the conflicting perspectives on the detrimental or beneficial roles of microglia. We previously showed that modulating the endocannabinoid tone by fatty acid amide hydrolase (FAAH) inactivation renders beneficial effects in an amyloidosis context, paradoxically accompanied by an exacerbated neuroinflammatory response and the enrichment of DAM population. Here, we aim to elucidate the role of microglial cells in FAAH-lacking mice in the 5xFAD mouse model of AD by using RNA-sequencing analysis, molecular determinations, and morphological studies by using in vivo multiphoton microscopy. FAAH-lacking AD mice displayed upregulated inflammatory genes and exhibited a DAM genetic profile. Conversely, genes linked to AD were downregulated. Depleting microglia using PLX5622 revealed that plaque-associated microglia in FAAH-deficient AD mice had a more stable, ramified morphology and increased Aβ uptake, leading to reduced plaque growth compared to control mice. Importantly, FAAH expression was negligible in microglial cells, thus suggesting a role for FAAH in the cellular interplay in the central nervous system. Our findings show that Faah gene inactivation triggers a hetero-cellular enhancement of microglial function that was paradoxically paralleled by an exacerbated inflammatory response. Taken together, the present data highlight FAAH as a potential therapeutic target in AD.
Collapse
Affiliation(s)
- María Andrea Arnanz
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - María Ferrer
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - María Teresa Grande
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | | | - Gonzalo Ruiz-Pérez
- Neuroscience Research Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, Department of Cell Biology, The Scripps Research Institute, San Diego, California, USA
- The Skaggs Institute for Chemical Biology, Department of Chemistry, The Scripps Research Institute, San Diego, California, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Víctor Javier Sánchez-Arévalo Lobo
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Julián Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | | |
Collapse
|
3
|
Rao A, Chen N, Kim MJ, Blumenfeld J, Yip O, Liang Z, Shostak D, Hao Y, Nelson MR, Koutsodendris N, Grone B, Ding L, Yoon SY, Arriola P, Zilberter M, Huang Y. Microglia depletion reduces human neuronal APOE4-related pathologies in a chimeric Alzheimer's disease model. Cell Stem Cell 2025; 32:86-104.e7. [PMID: 39500314 DOI: 10.1016/j.stem.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 11/13/2024]
Abstract
Despite strong evidence supporting the important roles of both apolipoprotein E4 (APOE4) and microglia in Alzheimer's disease (AD) pathogenesis, the effects of microglia on neuronal APOE4-related AD pathogenesis remain elusive. To examine such effects, we utilized microglial depletion in a chimeric model with induced pluripotent stem cell (iPSC)-derived human neurons in mouse hippocampus. Specifically, we transplanted homozygous APOE4, isogenic APOE3, and APOE-knockout (APOE-KO) iPSC-derived human neurons into the hippocampus of human APOE3 or APOE4 knockin mice and then depleted microglia in half of the chimeric mice. We found that both neuronal APOE and microglial presence were important for the formation of Aβ and tau pathologies in an APOE isoform-dependent manner (APOE4 > APOE3). Single-cell RNA sequencing analysis identified two pro-inflammatory microglial subtypes with elevated MHC-II gene expression enriched in chimeric mice with human APOE4 neuron transplants. These findings highlight the concerted roles of neuronal APOE, especially APOE4, and microglia in AD pathogenesis.
Collapse
Affiliation(s)
- Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nuo Chen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Zherui Liang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - David Shostak
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Leo Ding
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Pan J, Zhong J, Geng J, Oberhauser J, Shi S, Wan J. Microglial Lyzl4 Facilitates β-Amyloid Clearance in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412184. [PMID: 39555667 PMCID: PMC11727385 DOI: 10.1002/advs.202412184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Indexed: 11/19/2024]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative condition characterized by the accumulation and deposition of amyloid-β (Aβ) aggregates in the brain. Despite a wealth of research on the toxicity of Aβ and its role in synaptic damage, the mechanisms facilitating Aβ clearance are not yet fully understood. However, microglia, the primary immune cells of the central nervous system, are known to maintain homeostasis through the phagocytic clearance of protein aggregates and cellular debris. In this study, RNA sequencing analysis and live cell functional screens are employed to uncover microglial genetic modifiers related to AD. Lyzl4 is identified, which encodes a c-type lysozyme-like enzyme primarily localized to microglial lysosomes, as a gene significantly upregulated in AD microglia with aging and propose that Lyzl4 upregulation acts as a positive regulator of Aβ clearance. Furthermore, it is found that Lyzl4 overexpression boosts Aβ clearance both in vitro and in vivo, underscoring its potential for mitigating Aβ burden. These novel insights position Lyzl4 as a promising therapeutic target for Alzheimer's disease, paving the way for further exploration into potential AD treatments.
Collapse
Affiliation(s)
- Jie Pan
- Department of PathologyStanford University School of MedicinePalo AltoCA94305USA
| | - Jie Zhong
- Shenzhen Key Laboratory for Neuronal Structural BiologyBiomedical Research InstituteShenzhen Peking University – The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdong Province518036China
- Department of Systems BiologySchool of Life SciencesSouthern University of Science and TechnologyShenzhenGuangdong Province518055China
| | - Ji Geng
- Department of PathologyStanford University School of MedicinePalo AltoCA94305USA
| | - Jane Oberhauser
- Department of PathologyStanford University School of MedicinePalo AltoCA94305USA
- Neuroscience Graduate ProgramUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Shihua Shi
- Friedrich Miescher Institute for Biomedical Research (FMI)Basel4056Switzerland
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural BiologyBiomedical Research InstituteShenzhen Peking University – The Hong Kong University of Science and Technology Medical CenterShenzhenGuangdong Province518036China
- Department of NeuroscienceSchool of Life SciencesSouthern University of Science and TechnologyShenzhenGuangdong Province518055China
| |
Collapse
|
5
|
Guan J, Wu P, Liu M, Jiang C, Meng X, Wu X, Lu M, Fan Y, Gan L. Egln3 expression in microglia enhances the neuroinflammatory responses in Alzheimer's disease. Brain Behav Immun 2024; 125:21-32. [PMID: 39701332 DOI: 10.1016/j.bbi.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/27/2024] [Accepted: 12/14/2024] [Indexed: 12/21/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive and behavioral abnormalities, is the most prevalent neurodegenerative disease worldwide. Neuroinflammation, which is induced by microglial activation, resulting in the expression of a multitude of inflammatory factors, is one of the principal characteristics of AD. Herein, we found that Egln3 is differentially expressed in microglia in the brains of AD mice. Egln3 is a member of the Egln family of proline hydroxylases, which regulates a variety of biological processes, including transcription, the cell cycle, and apoptosis, through hydroxylation, ubiquitylation, and participation in glycolysis. To further observe the effects of Egln3 on cognitive function, we utilized APP/PS1 mice as a pathological model of AD to conduct behavioral experiments and assess the expression levels of Aβ and inflammatory factors. The specific mechanisms by which Egln3 affects microglial activation were analyzed using in vitro experiments and transcriptome sequencing. The results of these analyses demonstrated that Egln3 is highly expressed in microglia in AD. Inhibition of Egln3 expression in the brains of APP/PS1 mice improves neuroinflammatory responses and cognitive function, indicating that a high expression of Egln3 promotes AD progression. Furthermore, our findings indicate that Egln3 could activate the MAPK pathway, which in turn contributes to the aggravation of neuroinflammation. Inhibition of the MAPK pathway results in attenuation of the pro-inflammatory state of microglia. Consequently, Egln3 may exacerbate neuroinflammation and promote AD progression via the MAPK pathway in microglia, making it a promising target for AD-related therapies.
Collapse
Affiliation(s)
- Jiaxin Guan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Pengfei Wu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu 233030, China
| | - Meiling Liu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Xiaowei Wu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Meijiao Lu
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Ying Fan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China
| | - Lu Gan
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
6
|
Hu Y, Tao W. Current perspectives on microglia-neuron communication in the central nervous system: Direct and indirect modes of interaction. J Adv Res 2024; 66:251-265. [PMID: 38195039 PMCID: PMC11674795 DOI: 10.1016/j.jare.2024.01.006] [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: 04/07/2023] [Revised: 10/05/2023] [Accepted: 01/06/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The incessant communication that takes place between microglia and neurons is essential the development, maintenance, and pathogenesis of the central nervous system (CNS). As mobile phagocytic cells, microglia serve a critical role in surveilling and scavenging the neuronal milieu to uphold homeostasis. AIM OF REVIEW This review aims to discuss the various mechanisms that govern the interaction between microglia and neurons, from the molecular to the organ system level, and to highlight the importance of these interactions in the development, maintenance, and pathogenesis of the CNS. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent research has revealed that microglia-neuron interaction is vital for regulating fundamental neuronal functions, such as synaptic pruning, axonal remodeling, and neurogenesis. The review will elucidate the intricate signaling pathways involved in these interactions, both direct and indirect, to provide a better understanding of the fundamental mechanisms of brain function. Furthermore, gaining insights into these signals could lead to the development of innovative therapies for neural disorders.
Collapse
Affiliation(s)
- Yue Hu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiwei Tao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
7
|
Zong T, Li N, Han F, Liu J, Deng M, Li V, Zhang M, Zhou Y, Yu M. Microglial depletion rescues spatial memory impairment caused by LPS administration in adult mice. PeerJ 2024; 12:e18552. [PMID: 39559328 PMCID: PMC11572354 DOI: 10.7717/peerj.18552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
Recent studies have highlighted the importance of microglia, the resident macrophages in the brain, in regulating cognitive functions such as learning and memory in both healthy and diseased states. However, there are conflicting results and the underlying mechanisms are not fully understood. In this study, we examined the effect of depleting adult microglia on spatial learning and memory under both physiological conditions and lipopolysaccharide (LPS)-induced neuroinflammation. Our results revealed that microglial depletion by PLX5622 caused mild spatial memory impairment in mice under physiological conditions; however, it prevented memory deficits induced by systemic LPS insult. Inactivating microglia through minocycline administration replicated the protective effect of microglial depletion on LPS-induced memory impairment. Furthermore, our study showed that PLX5622 treatment suppressed LPS-induced neuroinflammation, microglial activation, and synaptic dysfunction. These results strengthen the evidence for the involvement of microglial immunoactivation in LPS-induced synaptic and cognitive malfunctions. They also suggest that targeting microglia may be a potential approach to treating neuroinflammation-associated cognitive dysfunction seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tao Zong
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Na Li
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Qingdao Binhai University, Qingdao, Shandong, China
| | - Fubing Han
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China, China
| | - Junru Liu
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Mingru Deng
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
| | - Vincent Li
- Beverly Hills High School, Unaffiliated, Beverly Hills, California, United States
| | - Meng Zhang
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
| | - Yu Zhou
- Affiliated Qingdao Third People’s Hospital, Department of Otorhinolaryngology Head and Neck, Qingdao University, Qingdao, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao, China, China
- Department of Neurology, Affiliated Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, China, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ming Yu
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
8
|
Quinn JP, Fisher K, Corbett N, Warwood S, Knight D, Kellett KA, Hooper NM. Proteolysis of tau by granzyme A in tauopathies generates fragments that are aggregation prone. Biochem J 2024; 481:1255-1274. [PMID: 39248243 PMCID: PMC11555691 DOI: 10.1042/bcj20240007] [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: 01/10/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 09/10/2024]
Abstract
Tauopathies, including Alzheimer's disease, corticobasal degeneration and progressive supranuclear palsy, are characterised by the aggregation of tau into insoluble neurofibrillary tangles in the brain. Tau is subject to a range of post-translational modifications, including proteolysis, that can promote its aggregation. Neuroinflammation is a hallmark of tauopathies and evidence is growing for a role of CD8+ T cells in disease pathogenesis. CD8+ T cells release granzyme proteases but what role these proteases play in neuronal dysfunction is currently lacking. Here, we identified that granzyme A (GzmA) is present in brain tissue and proteolytically cleaves tau. Mass spectrometric analysis of tau fragments produced on digestion of tau with GzmA identified three cleavage sites at R194-S195, R209-S210 and K240-S241. Mutation of the critical Arg or Lys residues at the cleavage sites in tau or chemical inhibition of GzmA blocked the proteolysis of tau by GzmA. Development of a semi-targeted mass spectrometry approach identified peptides in tauopathy brain tissue corresponding to proteolysis by GzmA at R209-S210 and K240-S241 in tau. When expressed in cells the GzmA-cleaved C-terminal fragments of tau were highly phosphorylated and aggregated upon incubation of the cells with tauopathy brain seed. The C-terminal fragment tau195-441 was able to transfer between cells and promote aggregation of tau in acceptor cells, indicating the propensity for such tau fragments to propagate between cells. Collectively, these results raise the possibility that GzmA, released from infiltrating cytotoxic CD8+ T cells, proteolytically cleaves tau into fragments that may contribute to its pathological properties in tauopathies.
Collapse
Affiliation(s)
- James P. Quinn
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Kate Fisher
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nicola Corbett
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Stacey Warwood
- Biological Mass Spectrometry Core Research Facility, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - David Knight
- Biological Mass Spectrometry Core Research Facility, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Katherine A.B. Kellett
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, U.K
| |
Collapse
|
9
|
Lu J, Zhang J, Wang X, Yuan F, Xin B, Li J, Yang Q, Li X, Zhang J, Wang X, Fu J, Guo C. Dl-3-n-butylphthalide promotes microglial phagocytosis and inhibits microglial inflammation via regulating AGE-RAGE pathway in APP/PS1 mice. Brain Res Bull 2024; 212:110969. [PMID: 38705540 DOI: 10.1016/j.brainresbull.2024.110969] [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: 02/05/2024] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Alzheimer's disease (AD) stands as the most prevalent neurodegenerative condition worldwide, and its correlation with microglial function is notably significant. Dl-3-n-butylphthalide (NBP), derived from the seeds of Apium graveolens L. (Chinese celery), has demonstrated the capacity to diminish Aβ levels in the brain tissue of Alzheimer's transgenic mice. Despite this, its connection to neuroinflammation and microglial phagocytosis, along with the specific molecular mechanism involved, remains undefined. In this study, NBP treatment exhibited a substantial improvement in learning deficits observed in AD transgenic mice (APP/PS1 transgenic mice). Furthermore, NBP treatment significantly mitigated the total cerebral Aβ plaque deposition. This effect was attributed to the heightened presence of activated microglia surrounding Aβ plaques and an increase in microglial phagocytosis of Aβ plaques. Transcriptome sequencing analysis unveiled the potential involvement of the AGE (advanced glycation end products) -RAGE (receptor for AGE) signaling pathway in NBP's impact on APP/PS1 mice. Subsequent investigation disclosed a reduction in the secretion of AGEs, RAGE, and proinflammatory factors within the hippocampus and cortex of NBP-treated APP/PS1 mice. In summary, NBP alleviates cognitive impairment by augmenting the number of activated microglia around Aβ plaques and ameliorating AGE-RAGE-mediated neuroinflammation. These findings underscore the related mechanism of the crucial neuroprotective roles of microglial phagocytosis and anti-inflammation in NBP treatment for AD, offering a potential therapeutic target for the disease.
Collapse
Affiliation(s)
- Jin Lu
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Division of Emergency Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Jiawei Zhang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China; Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Yuan
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Xin
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Li
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quanjun Yang
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingxia Li
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianping Zhang
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingyan Wang
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Jianliang Fu
- Department of Neurology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cheng Guo
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Bradford BM, Walmsley-Rowe L, Reynolds J, Verity N, Mabbott NA. Cell adhesion molecule CD44 is dispensable for reactive astrocyte activation during prion disease. Sci Rep 2024; 14:13749. [PMID: 38877012 PMCID: PMC11178777 DOI: 10.1038/s41598-024-63464-3] [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: 02/22/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
Prion diseases are fatal, infectious, neurodegenerative disorders resulting from accumulation of misfolded cellular prion protein in the brain. Early pathological changes during CNS prion disease also include reactive astrocyte activation with increased CD44 expression, microgliosis, as well as loss of dendritic spines and synapses. CD44 is a multifunctional cell surface adhesion and signalling molecule which is considered to play roles in astrocyte morphology and the maintenance of dendritic spine integrity and synaptic plasticity. However, the role of CD44 in prion disease was unknown. Here we used mice deficient in CD44 to determine the role of CD44 during prion disease. We show that CD44-deficient mice displayed no difference in their response to CNS prion infection when compared to wild type mice. Furthermore, the reactive astrocyte activation and microgliosis that accompanies CNS prion infection was unimpaired in the absence of CD44. Together, our data show that although CD44 expression is upregulated in reactive astrocytes during CNS prion disease, it is dispensable for astrocyte and microglial activation and the development of prion neuropathogenesis.
Collapse
Affiliation(s)
- Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| | - Lauryn Walmsley-Rowe
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Joe Reynolds
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
- Maurice Wohl Basic and Clinical Neuroscience Institute, King's College London, Denmark Hill, London, SE5 9NU, UK
| | - Nicholas Verity
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| |
Collapse
|
11
|
Seasons GM, Pellow C, Kuipers HF, Pike GB. Ultrasound and neuroinflammation: immune modulation via the heat shock response. Theranostics 2024; 14:3150-3177. [PMID: 38855178 PMCID: PMC11155413 DOI: 10.7150/thno.96270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Current pharmacological therapeutic approaches targeting chronic inflammation exhibit transient efficacy, often with adverse effects, limiting their widespread use - especially in the context of neuroinflammation. Effective interventions require the consideration of homeostatic function, pathway dysregulation, and pleiotropic effects when evaluating therapeutic targets. Signalling molecules have multiple functions dependent on the immune context, and this complexity results in therapeutics targeting a single signalling molecule often failing in clinical translation. Additionally, the administration of non-physiologic levels of neurotrophic or anti-inflammatory factors can alter endogenous signalling, resulting in unanticipated effects. Exacerbating these challenges, the central nervous system (CNS) is isolated by the blood brain barrier (BBB), restricting the infiltration of many pharmaceutical compounds into the brain tissue. Consequently, there has been marked interest in therapeutic techniques capable of modulating the immune response in a pleiotropic manner; ultrasound remains on this frontier. While ultrasound has been used therapeutically in peripheral tissues - accelerating healing in wounds, bone fractures, and reducing inflammation - it is only recently that it has been applied to the CNS. The transcranial application of low intensity pulsed ultrasound (LIPUS) has successfully mitigated neuroinflammation in vivo, in models of neurodegenerative disease across a broad spectrum of ultrasound parameters. To date, the underlying biological effects and signalling pathways modulated by ultrasound are poorly understood, with a diverse array of reported molecules implicated. The distributed nature of the beneficial response to LIPUS implies the involvement of an, as yet, undetermined upstream signalling pathway, homologous to the protective effect of febrile range hyperthermia in chronic inflammation. As such, we review the heat shock response (HSR), a protective signalling pathway activated by thermal and mechanical stress, as the possible upstream regulator of the anti-inflammatory effects of ultrasound.
Collapse
Affiliation(s)
- Graham M. Seasons
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Carly Pellow
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Hedwich F. Kuipers
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, University of Calgary, Alberta, T2N 1N4, Canada
| | - G. Bruce Pike
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
12
|
Bobotis BC, Halvorson T, Carrier M, Tremblay MÈ. Established and emerging techniques for the study of microglia: visualization, depletion, and fate mapping. Front Cell Neurosci 2024; 18:1317125. [PMID: 38425429 PMCID: PMC10902073 DOI: 10.3389/fncel.2024.1317125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
The central nervous system (CNS) is an essential hub for neuronal communication. As a major component of the CNS, glial cells are vital in the maintenance and regulation of neuronal network dynamics. Research on microglia, the resident innate immune cells of the CNS, has advanced considerably in recent years, and our understanding of their diverse functions continues to grow. Microglia play critical roles in the formation and regulation of neuronal synapses, myelination, responses to injury, neurogenesis, inflammation, and many other physiological processes. In parallel with advances in microglial biology, cutting-edge techniques for the characterization of microglial properties have emerged with increasing depth and precision. Labeling tools and reporter models are important for the study of microglial morphology, ultrastructure, and dynamics, but also for microglial isolation, which is required to glean key phenotypic information through single-cell transcriptomics and other emerging approaches. Strategies for selective microglial depletion and modulation can provide novel insights into microglia-targeted treatment strategies in models of neuropsychiatric and neurodegenerative conditions, cancer, and autoimmunity. Finally, fate mapping has emerged as an important tool to answer fundamental questions about microglial biology, including their origin, migration, and proliferation throughout the lifetime of an organism. This review aims to provide a comprehensive discussion of these established and emerging techniques, with applications to the study of microglia in development, homeostasis, and CNS pathologies.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
| | - Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec City, QC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
13
|
Sola-Sevilla N, Puerta E. SIRT2 as a potential new therapeutic target for Alzheimer's disease. Neural Regen Res 2024; 19:124-131. [PMID: 37488853 PMCID: PMC10479864 DOI: 10.4103/1673-5374.375315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/09/2023] [Accepted: 04/04/2023] [Indexed: 07/26/2023] Open
Abstract
Alzheimer's disease is the most common cause of dementia globally with an increasing incidence over the years, bringing a heavy burden to individuals and society due to the lack of an effective treatment. In this context, sirtuin 2, the sirtuin with the highest expression in the brain, has emerged as a potential therapeutic target for neurodegenerative diseases. This review summarizes and discusses the complex roles of sirtuin 2 in different molecular mechanisms involved in Alzheimer's disease such as amyloid and tau pathology, microtubule stability, neuroinflammation, myelin formation, autophagy, and oxidative stress. The role of sirtuin 2 in all these processes highlights its potential implication in the etiology and development of Alzheimer's disease. However, its presence in different cell types and its enormous variety of substrates leads to apparently contradictory conclusions when it comes to understanding its specific functions. Further studies in sirtuin 2 research with selective sirtuin 2 modulators targeting specific sirtuin 2 substrates are necessary to clarify its specific functions under different conditions and to validate it as a novel pharmacological target. This will contribute to the development of new treatment strategies, not only for Alzheimer's disease but also for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Noemi Sola-Sevilla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Elena Puerta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| |
Collapse
|
14
|
Phadke MS, Li J, Chen Z, Rodriguez PC, Mandula JK, Karapetyan L, Forsyth PA, Chen YA, Smalley KSM. Differential requirements for CD4+ T cells in the efficacy of the anti-PD-1+LAG-3 and anti-PD-1+CTLA-4 combinations in melanoma flank and brain metastasis models. J Immunother Cancer 2023; 11:e007239. [PMID: 38056899 PMCID: PMC10711842 DOI: 10.1136/jitc-2023-007239] [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] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Although the anti-PD-1+LAG-3 and the anti-PD-1+CTLA-4 combinations are effective in advanced melanoma, it remains unclear whether their mechanisms of action overlap. METHODS We used single cell (sc) RNA-seq, flow cytometry and IHC analysis of responding SM1, D4M-UV2 and B16 melanoma flank tumors and SM1 brain metastases to explore the mechanism of action of the anti-PD-1+LAG-3 and the anti-PD-1+CTLA-4 combination. CD4+ and CD8+ T cell depletion, tetramer binding assays and ELISPOT assays were used to demonstrate the unique role of CD4+T cell help in the antitumor effects of the anti-PD-1+LAG-3 combination. RESULTS The anti-PD-1+CTLA-4 combination was associated with the infiltration of FOXP3+regulatory CD4+ cells (Tregs), fewer activated CD4+T cells and the accumulation of a subset of IFNγ secreting cytotoxic CD8+T cells, whereas the anti-PD-1+LAG-3 combination led to the accumulation of CD4+T helper cells that expressed CXCR4, TNFSF8, IL21R and a subset of CD8+T cells with reduced expression of cytotoxic markers. T cell depletion studies showed a requirement for CD4+T cells for the anti-PD-1+LAG-3 combination, but not the PD-1-CTLA-4 combination at both flank and brain tumor sites. In anti-PD-1+LAG-3 treated tumors, CD4+T cell depletion was associated with fewer activated (CD69+) CD8+T cells and impaired IFNγ release but, conversely, increased numbers of activated CD8+T cells and IFNγ release in anti-PD-1+CTLA-4 treated tumors. CONCLUSIONS Together these studies suggest that these two clinically relevant immune checkpoint inhibitor (ICI) combinations have differential effects on CD4+T cell polarization, which in turn, impacted cytotoxic CD8+T cell function. Further insights into the mechanisms of action/resistance of these clinically-relevant ICI combinations will allow therapy to be further personalized.
Collapse
Affiliation(s)
- Manali S Phadke
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jiannong Li
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Cancer Center and Research Institute, Tampa, FL, USA
| | - Zhihua Chen
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Cancer Center and Research Institute, Tampa, FL, USA
| | - Paulo C Rodriguez
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jessica K Mandula
- Department of Immunology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lilit Karapetyan
- Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Peter A Forsyth
- Department of Neurooncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Y Ann Chen
- Department of Bioinformatics and Biostatistics, Moffitt Cancer Cancer Center and Research Institute, Tampa, FL, USA
| | - Keiran S M Smalley
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
15
|
Liang Y, Kang X, Zhang H, Xu H, Wu X. Knockdown and inhibition of hippocampal GPR17 attenuates lipopolysaccharide-induced cognitive impairment in mice. J Neuroinflammation 2023; 20:271. [PMID: 37990234 PMCID: PMC10662506 DOI: 10.1186/s12974-023-02958-9] [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: 08/11/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Previously we reported that inhibition of GPR17 prevents amyloid β 1-42 (Aβ1-42)-induced cognitive impairment in mice. However, the role of GPR17 on cognition is still largely unknown. METHODS Herein, we used a mouse model of cognitive impairment induced by lipopolysaccharide (LPS) to further investigate the role of GPR17 in cognition and its potential mechanism. The mice were pretreated with GPR17 shRNA lentivirus and cangrelor by microinjection into the dentate gyrus (DG) region of the hippocampus. After 21 days, LPS (0.25 mg/kg, i.p.) was administered for 7 days. Animal behavioral tests as well as pathological and biochemical assays were performed to evaluate the cognitive function in mice. RESULTS LPS exposure resulted in a significant increase in GPR17 expression at both protein and mRNA levels in the hippocampus. Gene reduction and pharmacological blockade of GPR17 improved cognitive impairment in both the Morris water maze and novel object recognition tests. Knockdown and inhibition of GPR17 inhibited Aβ production, decreased the expression of NF-κB p65, increased CREB phosphorylation and elevated BDNF expression, suppressed the accumulation of pro-inflammatory cytokines, inhibited Glial cells (microglia and astrocytes) activation, and increased Bcl-2, PSD-95, and SYN expression, reduced Bax expression as well as decreased caspase-3 activity and TUNEL-positive cells in the hippocampus of LPS-treated mice. Notably, knockdown and inhibition of GPR17 not only provided protective effects against cholinergic dysfunction but also facilitated the regulation of oxidative stress. In addition, cangrelor pretreatment can effectively inhibit the expression of inflammatory cytokines by suppressing NF-κB/CREB/BDNF signaling in BV-2 cells stimulated by LPS. However, activation of hippocampal GPR17 with MDL-29951 induced cognitive impairment in normal mice. CONCLUSIONS These observations indicate that GPR17 may possess a neuroprotective effect against LPS-induced cognition deficits, and neuroinflammation by modulation of NF-κB/CREB/BDNF signaling in mice, indicating that GPR17 may be a promising new target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yusheng Liang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xu Kang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Haiwang Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Heng Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China
| | - Xian Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
16
|
Gaunt JR, Zainolabidin N, Yip AKK, Tan JM, Low AYT, Chen AI, Ch'ng TH. Cytokine enrichment in deep cerebellar nuclei is contributed by multiple glial populations and linked to reduced amyloid plaque pathology. J Neuroinflammation 2023; 20:269. [PMID: 37978387 PMCID: PMC10656954 DOI: 10.1186/s12974-023-02913-8] [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: 04/24/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease (AD) pathology and amyloid-beta (Aβ) plaque deposition progress slowly in the cerebellum compared to other brain regions, while the entorhinal cortex (EC) is one of the most vulnerable regions. Using a knock-in AD mouse model (App KI), we show that within the cerebellum, the deep cerebellar nuclei (DCN) has particularly low accumulation of Aβ plaques. To identify factors that might underlie differences in the progression of AD-associated neuropathology across regions, we profiled gene expression in single nuclei (snRNAseq) across all cell types in the DCN and EC of wild-type (WT) and App KI male mice at age 7 months. We found differences in expression of genes associated with inflammatory activation, PI3K-AKT signalling, and neuron support functions between both regions and genotypes. In WT mice, the expression of interferon-response genes in microglia is higher in the DCN than the EC and this enrichment is confirmed by RNA in situ hybridisation, and measurement of inflammatory cytokines by protein array. Our analyses also revealed that multiple glial populations are responsible for establishing this cytokine-enriched niche. Furthermore, homogenates derived from the DCN induced inflammatory gene expression in BV2 microglia. We also assessed the relationship between the DCN microenvironment and Aβ pathology by depleting microglia using a CSF1R inhibitor PLX5622 and saw that, surprisingly, the expression of a subset of inflammatory cytokines was increased while plaque abundance in the DCN was further reduced. Overall, our study revealed the presence of a cytokine-enriched microenvironment unique to the DCN that when modulated, can alter plaque deposition.
Collapse
Affiliation(s)
- Jessica R Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Norliyana Zainolabidin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Alaric K K Yip
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Aloysius Y T Low
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Albert I Chen
- Center for Aging Research, Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA, 92121, USA.
- Molecular Neurobiology Laboratory, Salk Institute, La Jolla, CA, 92037, USA.
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, Singapore, 308232, Singapore.
- School of Biological Science, Nanyang Technological University, Singapore, 63755, Singapore.
| |
Collapse
|
17
|
Rao A, Chen N, Kim MJ, Blumenfeld J, Yip O, Hao Y, Liang Z, Nelson MR, Koutsodendris N, Grone B, Ding L, Yoon SY, Arriola P, Huang Y. Microglia Depletion Reduces Human Neuronal APOE4-Driven Pathologies in a Chimeric Alzheimer's Disease Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566510. [PMID: 38014339 PMCID: PMC10680610 DOI: 10.1101/2023.11.10.566510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Despite strong evidence supporting the involvement of both apolipoprotein E4 (APOE4) and microglia in Alzheimer's Disease (AD) pathogenesis, the effects of microglia on neuronal APOE4-driven AD pathogenesis remain elusive. Here, we examined such effects utilizing microglial depletion in a chimeric model with human neurons in mouse hippocampus. Specifically, we transplanted homozygous APOE4, isogenic APOE3, and APOE-knockout (APOE-KO) induced pluripotent stem cell (iPSC)-derived human neurons into the hippocampus of human APOE3 or APOE4 knock-in mice, and depleted microglia in half the chimeric mice. We found that both neuronal APOE and microglial presence were important for the formation of Aβ and tau pathologies in an APOE isoform-dependent manner (APOE4 > APOE3). Single-cell RNA-sequencing analysis identified two pro-inflammatory microglial subtypes with high MHC-II gene expression that are enriched in chimeric mice with human APOE4 neuron transplants. These findings highlight the concerted roles of neuronal APOE, especially APOE4, and microglia in AD pathogenesis.
Collapse
Affiliation(s)
- Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Nuo Chen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Zherui Liang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
| | - Maxine R. Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Leo Ding
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, USA
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Afsar A, Chen M, Xuan Z, Zhang L. A glance through the effects of CD4 + T cells, CD8 + T cells, and cytokines on Alzheimer's disease. Comput Struct Biotechnol J 2023; 21:5662-5675. [PMID: 38053545 PMCID: PMC10694609 DOI: 10.1016/j.csbj.2023.10.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Unfortunately, despite numerous studies, an effective treatment for AD has not yet been established. There is remarkable evidence indicating that the innate immune mechanism and adaptive immune response play significant roles in the pathogenesis of AD. Several studies have reported changes in CD8+ and CD4+ T cells in AD patients. This mini-review article discusses the potential contribution of CD4+ and CD8+ T cells reactivity to amyloid β (Aβ) protein in individuals with AD. Moreover, this mini-review examines the potential associations between T cells, heme oxygenase (HO), and impaired mitochondria in the context of AD. While current mathematical models of AD have not extensively addressed the inclusion of CD4+ and CD8+ T cells, there exist models that can be extended to consider AD as an autoimmune disease involving these T cell types. Additionally, the mini-review covers recent research that has investigated the utilization of machine learning models, considering the impact of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Atefeh Afsar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Min Chen
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
19
|
Palsamy K, Chen JY, Skaggs K, Qadeer Y, Connors M, Cutler N, Richmond J, Kommidi V, Poles A, Affrunti D, Powell C, Goldman D, Parent JM. Microglial depletion after brain injury prolongs inflammation and impairs brain repair, adult neurogenesis and pro-regenerative signaling. Glia 2023; 71:2642-2663. [PMID: 37449457 PMCID: PMC10528132 DOI: 10.1002/glia.24444] [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: 02/16/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The adult zebrafish brain, unlike mammals, has a remarkable regenerative capacity. Although inflammation in part hinders regeneration in mammals, it is necessary for zebrafish brain repair. Microglia are resident brain immune cells that regulate the inflammatory response. To explore the microglial role in repair, we used liposomal clodronate or colony stimulating factor-1 receptor (csf1r) inhibitor to suppress microglia after brain injury, and also examined regeneration in two genetic mutant lines that lack microglia. We found that microglial ablation impaired telencephalic regeneration after injury. Microglial suppression attenuated cell proliferation at the intermediate progenitor cell amplification stage of neurogenesis. Notably, the loss of microglia impaired phospho-Stat3 (signal transducer and activator of transcription 3) and ß-Catenin signaling after injury. Furthermore, the ectopic activation of Stat3 and ß-Catenin rescued neurogenesis defects caused by microglial loss. Microglial suppression also prolonged the post-injury inflammatory phase characterized by neutrophil accumulation, likely hindering the resolution of inflammation. These findings reveal specific roles of microglia and inflammatory signaling during zebrafish telencephalic regeneration that should advance strategies to improve mammalian brain repair.
Collapse
Affiliation(s)
- Kanagaraj Palsamy
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica Y Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kaia Skaggs
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- University of Findlay, Findlay, Ohio, USA
| | - Yusuf Qadeer
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Meghan Connors
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Cutler
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joshua Richmond
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Vineeth Kommidi
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison Poles
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Danielle Affrunti
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Curtis Powell
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Goldman
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Yang H, Qin Q, Wang M, Yin Y, Li R, Tang Y. Crosstalk between peripheral immunity and central nervous system in Alzheimer's disease. Cell Immunol 2023; 391-392:104743. [PMID: 37451918 DOI: 10.1016/j.cellimm.2023.104743] [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: 06/28/2022] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The significance of peripheral immunity in the pathogenesis and progression of Alzheimer's diseases (AD) has been recognized. Brain-infiltrated peripheral immune components transporting across the blood-brain barrier (BBB) may reshape the central immune environment. However, mechanisms of how these components open the BBB for AD occurrence and development and correlations between peripheral and central immunity have not been fully explored. Herein, we formulate a hypothesis whereby peripheral immunity as a critical factor allows AD to progress. Peripheral central immune cell crosstalk is associated with early AD pathology and related risk factors. The damaged BBB permits peripheral immune cells to enter the central immune system to deprive its immune privilege promoting the progression toward developing AD. This review summarizes the influences of risk factors on peripheral immunity, alongside their functions, highlighting the concept of peripheral and central immunity as an integrated system in AD pathogenesis, which has received scant attention before.
Collapse
Affiliation(s)
- Hanchen Yang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Meng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yunsi Yin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ruiyang Li
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China.
| |
Collapse
|
21
|
Lang H, Noble KV, Barth JL, Rumschlag JA, Jenkins TR, Storm SL, Eckert MA, Dubno JR, Schulte BA. The Stria Vascularis in Mice and Humans Is an Early Site of Age-Related Cochlear Degeneration, Macrophage Dysfunction, and Inflammation. J Neurosci 2023; 43:5057-5075. [PMID: 37268417 PMCID: PMC10324995 DOI: 10.1523/jneurosci.2234-22.2023] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/19/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023] Open
Abstract
Age-related hearing loss, or presbyacusis, is a common degenerative disorder affecting communication and quality of life for millions of older adults. Multiple pathophysiologic manifestations, along with many cellular and molecular alterations, have been linked to presbyacusis; however, the initial events and causal factors have not been clearly established. Comparisons of the transcriptome in the lateral wall (LW) with other cochlear regions in a mouse model (of both sexes) of "normal" age-related hearing loss revealed that early pathophysiological alterations in the stria vascularis (SV) are associated with increased macrophage activation and a molecular signature indicative of inflammaging, a common form of immune dysfunction. Structure-function correlation analyses in mice across the lifespan showed that the age-dependent increase in macrophage activation in the stria vascularis is associated with a decline in auditory sensitivity. High-resolution imaging analysis of macrophage activation in middle-aged and aged mouse and human cochleas, along with transcriptomic analysis of age-dependent changes in mouse cochlear macrophage gene expression, support the hypothesis that aberrant macrophage activity is an important contributor to age-dependent strial dysfunction, cochlear pathology, and hearing loss. Thus, this study highlights the SV as a primary site of age-related cochlear degeneration and aberrant macrophage activity and dysregulation of the immune system as early indicators of age-related cochlear pathology and hearing loss. Importantly, novel new imaging methods described here now provide a means to analyze human temporal bones in a way that had not previously been feasible and thereby represent a significant new tool for otopathological evaluation.SIGNIFICANCE STATEMENT Age-related hearing loss is a common neurodegenerative disorder affecting communication and quality of life. Current interventions (primarily hearing aids and cochlear implants) offer imperfect and often unsuccessful therapeutic outcomes. Identification of early pathology and causal factors is crucial for the development of new treatments and early diagnostic tests. Here, we find that the SV, a nonsensory component of the cochlea, is an early site of structural and functional pathology in mice and humans that is characterized by aberrant immune cell activity. We also establish a new technique for evaluating cochleas from human temporal bones, an important but understudied area of research because of a lack of well-preserved human specimens and difficult tissue preparation and processing approaches.
Collapse
Affiliation(s)
- Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kenyaria V Noble
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeremy L Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jeffrey A Rumschlag
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Tyreek R Jenkins
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Shelby L Storm
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mark A Eckert
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Judy R Dubno
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Bradley A Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
22
|
de Sousa DMB, Poupardin R, Villeda SA, Schroer AB, Fröhlich T, Frey V, Staffen W, Mrowetz H, Altendorfer B, Unger MS, Iglseder B, Paulweber B, Trinka E, Cadamuro J, Drerup M, Schallmoser K, Aigner L, Kniewallner KM. The platelet transcriptome and proteome in Alzheimer's disease and aging: an exploratory cross-sectional study. Front Mol Biosci 2023; 10:1196083. [PMID: 37457829 PMCID: PMC10348715 DOI: 10.3389/fmolb.2023.1196083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: Alzheimer's disease (AD) and aging are associated with platelet hyperactivity. However, the mechanisms underlying abnormal platelet function in AD and aging are yet poorly understood. Methods: To explore the molecular profile of AD and aged platelets, we investigated platelet activation (i.e., CD62P expression), proteome and transcriptome in AD patients, non-demented elderly, and young individuals as controls. Results: AD, aged and young individuals showed similar levels of platelet activation based on CD62P expression. However, AD and aged individuals had a proteomic signature suggestive of increased platelet activation compared with young controls. Transcriptomic profiling suggested the dysregulation of proteolytic machinery involved in regulating platelet function, particularly the ubiquitin-proteasome system in AD and autophagy in aging. The functional implication of these transcriptomic alterations remains unclear and requires further investigation. Discussion: Our data strengthen the evidence of enhanced platelet activation in aging and provide a first glimpse of the platelet transcriptomic changes occurring in AD.
Collapse
Affiliation(s)
- Diana M. Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Rodolphe Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Saul A. Villeda
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Adam B. Schroer
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Thomas Fröhlich
- Laboratory of Functional Genome Analysis (LAFUGA), Gene Center, Ludwig Maximilian University of Munich, Munich, Germany
| | - Vanessa Frey
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Department of Neurology, Christian Doppler Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Wolfgang Staffen
- Department of Neurology, Christian Doppler Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Michael S. Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Iglseder
- Department of Neurology, Christian Doppler Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Paulweber
- Department of Internal Medicine, St. Johanns University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler Clinic, Paracelsus Medical University, Salzburg, Austria
- Department of Public Health, Health Services Research and Health Technology Assessment, UMIT-University for Health Sciences, Medical Informatics and Technology, Hall in Tirol, Austria
- Neuroscience Institute, Christian Doppler University Hospital, Paracelsus Medical University and Centre for Cognitive Neuroscience Salzburg, Salzburg, Austria
| | - Janne Cadamuro
- Department of Laboratory Medicine, University Hospital SALK, Salzburg, Austria
| | - Martin Drerup
- Department of Urology, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Department of Transfusion Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Kathrin M. Kniewallner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
23
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [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: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
24
|
Wang YY, Zhou YN, Jiang L, Wang S, Zhu L, Zhang SS, Yang H, He Q, Liu L, Xie YH, Liang X, Tang J, Chao FL, Tang Y. Long-term voluntary exercise inhibited AGE/RAGE and microglial activation and reduced the loss of dendritic spines in the hippocampi of APP/PS1 transgenic mice. Exp Neurol 2023; 363:114371. [PMID: 36871860 DOI: 10.1016/j.expneurol.2023.114371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Alzheimer's disease (AD) is closely related to hippocampal synapse loss, which can be alleviated by running exercise. However, further studies are needed to determine whether running exercise reduces synapse loss in the hippocampus in an AD model by regulating microglia. Ten-month-old male wild-type mice and APP/PS1 mice were randomly divided into control and running groups. All mice in the running groups were subjected to voluntary running exercise for four months. After the behavioral tests, immunohistochemistry, stereological methods, immunofluorescence staining, 3D reconstruction, western blotting and RNA-Seq were performed. Running exercise improved the spatial learning and memory abilities of APP/PS1 mice and increased the total number of dendritic spines, the levels of the PSD-95 and Synapsin Ia/b proteins, the colocalization of PSD-95 and neuronal dendrites (MAP-2) and the number of PSD-95-contacting astrocytes (GFAP) in the hippocampi of APP/PS1 mice. Moreover, running exercise reduced the relative expression of CD68 and Iba-1, the number of Iba-1+ microglia and the colocalization of PSD-95 and Iba-1+ microglia in the hippocampi of APP/PS1 mice. The RNA-Seq results showed that some differentially expressed genes (DEGs) related to the complement system (Cd59b, Serping1, Cfh, A2m, and Trem2) were upregulated in the hippocampi of APP/PS1 mice, while running exercise downregulated the C3 gene. At the protein level, running exercise also reduced the expression of advanced glycation end products (AGEs), receptor for advanced glycation end products (RAGE), C1q and C3 in the hippocampus and AGEs and RAGE in hippocampal microglia in APP/PS1 mice. Furthermore, the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes were upregulated in the hippocampi of APP/PS1 mice but downregulated after running, and these genes were associated with the C3 and RAGE genes according to protein-protein interaction (PPI) analysis. These findings indicate that long-term voluntary exercise might protect hippocampal synapses and affect the function and activation of microglia, the AGE/RAGE signaling pathway in microglia and the C1q/C3 complement system in the hippocampus in APP/PS1 mice, and these effects may be related to the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes. The current results provide an important basis for identifying targets for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yi-Ying Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Ning Zhou
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Shun Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zhu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan-Shan Zhang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Li Liu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Han Xie
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Department of Pathophysiology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
25
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
26
|
de Sousa DMB, Benedetti A, Altendorfer B, Mrowetz H, Unger MS, Schallmoser K, Aigner L, Kniewallner KM. Immune-mediated platelet depletion augments Alzheimer's disease neuropathological hallmarks in APP-PS1 mice. Aging (Albany NY) 2023; 15:630-649. [PMID: 36734880 PMCID: PMC9970308 DOI: 10.18632/aging.204502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/23/2023] [Indexed: 02/04/2023]
Abstract
In Alzheimer's disease (AD), platelets become dysfunctional and might contribute to amyloid beta deposition. Here, we depleted platelets in one-year-old APP Swedish PS1 dE9 (APP-PS1) transgenic mice for five days, using intraperitoneal injections of an anti-CD42b antibody, and assessed changes in cerebral amyloidosis, plaque-associated neuritic dystrophy and gliosis. In APP-PS1 female mice, platelet depletion shifted amyloid plaque size distribution towards bigger plaques and increased neuritic dystrophy in the hippocampus. In platelet-depleted females, plaque-associated Iba1+ microglia had lower amounts of fibrillar amyloid beta cargo and GFAP+ astrocytic processes showed a higher overlap with thioflavin S+ amyloid plaques. In contrast to the popular hypothesis that platelets foster plaque pathology, our data suggest that platelets might limit plaque growth and attenuate plaque-related neuritic dystrophy at advanced stages of amyloid plaque pathology in APP-PS1 female mice. Whether the changes in amyloid plaque pathology are due to a direct effect on amyloid beta deposition or are a consequence of altered glial function needs to be further elucidated.
Collapse
Affiliation(s)
- Diana M. Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Ariane Benedetti
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria,Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Michael S. Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Katharina Schallmoser
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria,Department of Transfusion Medicine, University Clinic, Paracelsus Medical University, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Kathrin Maria Kniewallner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
27
|
Basilico B, Ferrucci L, Khan A, Di Angelantonio S, Ragozzino D, Reverte I. What microglia depletion approaches tell us about the role of microglia on synaptic function and behavior. Front Cell Neurosci 2022; 16:1022431. [PMID: 36406752 PMCID: PMC9673171 DOI: 10.3389/fncel.2022.1022431] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Microglia are dynamic cells, constantly surveying their surroundings and interacting with neurons and synapses. Indeed, a wealth of knowledge has revealed a critical role of microglia in modulating synaptic transmission and plasticity in the developing brain. In the past decade, novel pharmacological and genetic strategies have allowed the acute removal of microglia, opening the possibility to explore and understand the role of microglia also in the adult brain. In this review, we summarized and discussed the contribution of microglia depletion strategies to the current understanding of the role of microglia on synaptic function, learning and memory, and behavior both in physiological and pathological conditions. We first described the available microglia depletion methods highlighting their main strengths and weaknesses. We then reviewed the impact of microglia depletion on structural and functional synaptic plasticity. Next, we focused our analysis on the effects of microglia depletion on behavior, including general locomotor activity, sensory perception, motor function, sociability, learning and memory both in healthy animals and animal models of disease. Finally, we integrated the findings from the reviewed studies and discussed the emerging roles of microglia on the maintenance of synaptic function, learning, memory strength and forgetfulness, and the implications of microglia depletion in models of brain disease.
Collapse
Affiliation(s)
| | - Laura Ferrucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Azka Khan
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Davide Ragozzino
- Laboratory Affiliated to Institute Pasteur Italia – Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- *Correspondence: Davide Ragozzino,
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- Ingrid Reverte,
| |
Collapse
|
28
|
Battis K, Florio JB, Mante M, Lana A, Naumann I, Gauer C, Lambrecht V, Müller SJ, Cobo I, Fixsen B, Kim HY, Masliah E, Glass CK, Schlachetzki JCM, Rissman RA, Winkler J, Hoffmann A. CSF1R-Mediated Myeloid Cell Depletion Prolongs Lifespan But Aggravates Distinct Motor Symptoms in a Model of Multiple System Atrophy. J Neurosci 2022; 42:7673-7688. [PMID: 36333098 PMCID: PMC9546481 DOI: 10.1523/jneurosci.0417-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/03/2022] [Accepted: 08/15/2022] [Indexed: 01/21/2023] Open
Abstract
As the CNS-resident macrophages and member of the myeloid lineage, microglia fulfill manifold functions important for brain development and homeostasis. In the context of neurodegenerative diseases, they have been implicated in degenerative and regenerative processes. The discovery of distinct activation patterns, including increased phagocytosis, indicated a damaging role of myeloid cells in multiple system atrophy (MSA), a devastating, rapidly progressing atypical parkinsonian disorder. Here, we analyzed the gene expression profile of microglia in a mouse model of MSA (MBP29-hα-syn) and identified a disease-associated expression profile and upregulation of the colony-stimulating factor 1 (Csf1). Thus, we hypothesized that CSF1 receptor-mediated depletion of myeloid cells using PLX5622 modifies the disease progression and neuropathological phenotype in this mouse model. Intriguingly, sex-balanced analysis of myeloid cell depletion in MBP29-hα-syn mice revealed a two-faced outcome comprising an improved survival rate accompanied by a delayed onset of neurological symptoms in contrast to severely impaired motor functions. Furthermore, PLX5622 reversed gene expression profiles related to myeloid cell activation but reduced gene expression associated with transsynaptic signaling and signal release. While transcriptional changes were accompanied by a reduction of dopaminergic neurons in the SNpc, striatal neuritic density was increased upon myeloid cell depletion in MBP29-hα-syn mice. Together, our findings provide insight into the complex, two-faced role of myeloid cells in the context of MSA emphasizing the importance to carefully balance the beneficial and adverse effects of CSF1R inhibition in different models of neurodegenerative disorders before its clinical translation.SIGNIFICANCE STATEMENT Myeloid cells have been implicated as detrimental in the disease pathogenesis of multiple system atrophy. However, long-term CSF1R-dependent depletion of these cells in a mouse model of multiple system atrophy demonstrates a two-faced effect involving an improved survival associated with a delayed onset of disease and reduced inflammation which was contrasted by severely impaired motor functions, synaptic signaling, and neuronal circuitries. Thus, this study unraveled a complex role of myeloid cells in multiple system atrophy, which indicates important functions beyond the previously described disease-associated, destructive phenotype and emphasized the need of further investigation to carefully and individually fine-tune immunologic processes in different neurodegenerative diseases.
Collapse
Affiliation(s)
- Kristina Battis
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Jazmin B Florio
- Department of Neurosciences, University of California-San Diego, La Jolla, California 92093
| | - Michael Mante
- Department of Neurosciences, University of California-San Diego, La Jolla, California 92093
| | - Addison Lana
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093
| | - Isabel Naumann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Carina Gauer
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Vera Lambrecht
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Simon Julian Müller
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Isidoro Cobo
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093
| | - Bethany Fixsen
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093
| | - Ha Yeon Kim
- Department of Neurosciences, University of California-San Diego, La Jolla, California 92093
| | - Eliezer Masliah
- Division of Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California-San Diego, La Jolla, California 92093
| | - Robert A Rissman
- Department of Neurosciences, University of California-San Diego, La Jolla, California 92093
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| | - Alana Hoffmann
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, 91054, Germany
| |
Collapse
|
29
|
Altendorfer B, Unger MS, Poupardin R, Hoog A, Asslaber D, Gratz IK, Mrowetz H, Benedetti A, de Sousa DMB, Greil R, Egle A, Gate D, Wyss-Coray T, Aigner L. Transcriptomic Profiling Identifies CD8 + T Cells in the Brain of Aged and Alzheimer's Disease Transgenic Mice as Tissue-Resident Memory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1272-1285. [PMID: 36165202 PMCID: PMC9515311 DOI: 10.4049/jimmunol.2100737] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/20/2022] [Indexed: 12/13/2022]
Abstract
Peripheral immune cell infiltration into the brain is a prominent feature in aging and various neurodegenerative diseases such as Alzheimer's disease (AD). As AD progresses, CD8+ T cells infiltrate into the brain parenchyma, where they tightly associate with neurons and microglia. The functional properties of CD8+ T cells in the brain are largely unknown. To gain further insights into the putative functions of CD8+ T cells in the brain, we explored and compared the transcriptomic profile of CD8+ T cells isolated from the brain and blood of transgenic AD (APPswe/PSEN1dE9, line 85 [APP-PS1]) and age-matched wild-type (WT) mice. Brain CD8+ T cells of APP-PS1 and WT animals had similar transcriptomic profiles and substantially differed from blood circulating CD8+ T cells. The gene signature of brain CD8+ T cells identified them as tissue-resident memory (Trm) T cells. Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis on the significantly upregulated genes revealed overrepresentation of biological processes involved in IFN-β signaling and the response to viral infections. Furthermore, brain CD8+ T cells of APP-PS1 and aged WT mice showed similar differentially regulated genes as brain Trm CD8+ T cells in mouse models with acute virus infection, chronic parasite infection, and tumor growth. In conclusion, our profiling of brain CD8+ T cells suggests that in AD, these cells exhibit similar adaptive immune responses as in other inflammatory diseases of the CNS, potentially opening the door for immunotherapy in AD.
Collapse
Affiliation(s)
- Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Michael Stefan Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Rodolphe Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Anna Hoog
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Daniela Asslaber
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Iris Karina Gratz
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Ariane Benedetti
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria
| | - Diana Marisa Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Richard Greil
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Alexander Egle
- IIIrd Medical Department with Hematology and Medical Oncology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - David Gate
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA; and
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA; and
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria;
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
30
|
Foray C, Barca C, Winkeler A, Wagner S, Hermann S, Schäfers M, Grauer OM, Zinnhardt B, Jacobs AH. Interrogating Glioma-Associated Microglia and Macrophage Dynamics Under CSF-1R Therapy with Multitracer In Vivo PET/MRI. J Nucl Med 2022; 63:1386-1393. [PMID: 35115369 PMCID: PMC9454459 DOI: 10.2967/jnumed.121.263318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/04/2022] [Indexed: 01/26/2023] Open
Abstract
Glioma-associated microglia and macrophages (GAMMs) are key players in creating an immunosuppressive microenvironment. They can be efficiently targeted by inhibiting the colony-stimulating factor 1 receptor (CSF-1R). We applied noninvasive PET/CT and PET/MRI using 18F-fluoroethyltyrosine (18F-FET) (amino acid metabolism) and N,N-diethyl-2-[4-(2-18F-fluoroethoxy)phenyl]-5,7-dimethylpyrazolo[1,5-a]pyrimidine-3-acetamide (18F-DPA-714) (translocator protein) to understand the role of GAMMs in glioma initiation, monitor in vivo therapy-induced GAMM depletion, and observe GAMM repopulation after drug withdrawal. Methods: C57BL/6 mice (n = 44) orthotopically implanted with syngeneic mouse GL261 glioma cells were treated with different regimens using the CSF-1R inhibitor PLX5622 (6-fluoro-N-((5-fluoro-2-methoxypyridin-3-yl)methyl)-5-((5-methyl-1H-pyrrolo[2,3-b]pyridin-3-yl)methyl)pyridin-2-amine) or vehicle, establishing a preconditioning model and a repopulation model, respectively. The mice underwent longitudinal PET/CT and PET/MRI. Results: The preconditioning model indicated similar tumor growth based on MRI (44.5% ± 24.8%), 18F-FET PET (18.3% ± 11.3%), and 18F-DPA-714 PET (16% ± 19.04%) volume dynamics in all groups, suggesting that GAMMs are not involved in glioma initiation. The repopulation model showed significantly reduced 18F-DPA-714 uptake (-45.6% ± 18.4%), significantly reduced GAMM infiltration even after repopulation, and a significantly decreased tumor volume (-54.29% ± 8.6%) with repopulation as measured by MRI, supported by a significant reduction in 18F-FET uptake (-50.2% ± 5.3%). Conclusion: 18F-FET and 18F-DPA-714 PET/MRI allow noninvasive assessment of glioma growth under various regimens of CSF-1R therapy. CSF-1R-mediated modulation of GAMMs may be of high interest as therapy or cotherapy against glioma.
Collapse
Affiliation(s)
- Claudia Foray
- European Institute for Molecular Imaging, University of Münster, Münster, Germany; .,PET Imaging in Drug Design and Development, Münster, Germany
| | - Cristina Barca
- European Institute for Molecular Imaging, University of Münster, Münster, Germany;,PET Imaging in Drug Design and Development, Münster, Germany
| | | | - Stefan Wagner
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Michael Schäfers
- European Institute for Molecular Imaging, University of Münster, Münster, Germany;,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Oliver M. Grauer
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Bastian Zinnhardt
- European Institute for Molecular Imaging, University of Münster, Münster, Germany;,PET Imaging in Drug Design and Development, Münster, Germany;,Department of Nuclear Medicine, University Hospital Münster, Münster, Germany;,Biomarkers and Translational Technologies, Neurosciences and Rare Diseases, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland; and
| | - Andreas H. Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany;,PET Imaging in Drug Design and Development, Münster, Germany;,Department of Geriatrics with Neurology, Johanniter Hospital, and Centre for Integrated Oncology of the University Hospital Bonn, Bonn, Germany
| |
Collapse
|
31
|
Wendt S, Johnson S, Weilinger NL, Groten C, Sorrentino S, Frew J, Yang L, Choi HB, Nygaard HB, MacVicar BA. Simultaneous imaging of redox states in dystrophic neurites and microglia at Aβ plaques indicate lysosome accumulation not microglia correlate with increased oxidative stress. Redox Biol 2022; 56:102448. [PMID: 36037587 PMCID: PMC9440309 DOI: 10.1016/j.redox.2022.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
The inter-relationship between microglia dynamics and oxidative stress (Ox-stress) in dystrophic neurites (DNs) at Alzheimer's Disease (AD) plaques may contribute to the pathological changes in neurons. We developed new in vivo imaging strategies to combine EGFP expression in microglia with neuronal expression of genetically encoded ratiometric redox sensors (rogRFP2 or roGFP1), and immunohistochemistry to investigate how microglia influence Ox-stress at amyloid plaques in 5xFAD AD mice. By simultaneously imaging microglia morphology and neuronal Ox-stress over time in vivo and in fixed brains we found that microglia preferentially enwrapped DNs exhibiting the greatest degree of Ox-stress. After microglia were partially depleted with the CSF1 receptor antagonist PLX3397, Ox-stress in DNs increased in a manner that was inversely correlated to the extent of coverage of the adjacent Aβ plaques by the remaining microglia. These data suggest that microglia do not create Ox-stress at Aβ plaques but instead create protective barriers around Aβ plaques possibly reducing the spread of Aβ. Intracranial injection of Aβ was sufficient to induce neuronal Ox-stress suggesting it to be the initial trigger of Ox-stress generation. Although Ox-stress is increased in DNs, neuronal survival is enhanced following microglia depletion indicating complex and multifactorial roles of microglia with both neurotoxic and neuroprotective components. Increased Ox-stress of DNs was correlated with higher LAMP1 and ubiquitin immunoreactivity supporting proposed mechanistic links between lysosomal accumulation in DNs and their intrinsic generation of Ox-stress. Our results suggest protective as well as neurotoxic roles for microglia at plaques and that the generation of Ox-stress of DNs could intrinsically be generated via lysosomal disruption rather than by microglia. In Brief: Simultaneous imaging of microglia and neuronal Ox-stress revealed a double-edged role for microglia in 5xFAD mice. Plaque associated microglia were attracted to and enwrapped Aβ plaques as well as the most highly oxidized DNs. After partial depletion of microglia, DNs were larger with greater levels of Ox-stress. Despite increased Ox-stress after microglia removal neuronal survival improved. Greater Ox-stress was correlated with increased levels of LAMP1 and ubiquitin thereby linking lysosome accumulation and Ox-stress in DNs.
Collapse
Affiliation(s)
- Stefan Wendt
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| | - Sora Johnson
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Nicholas L Weilinger
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Christopher Groten
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Stefano Sorrentino
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Jonathan Frew
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Lucy Yang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Hyun B Choi
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Haakon B Nygaard
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Brian A MacVicar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
32
|
St-Pierre MK, VanderZwaag J, Loewen S, Tremblay MÈ. All roads lead to heterogeneity: The complex involvement of astrocytes and microglia in the pathogenesis of Alzheimer’s disease. Front Cell Neurosci 2022; 16:932572. [PMID: 36035256 PMCID: PMC9413962 DOI: 10.3389/fncel.2022.932572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, glial cells have been acknowledged as key players in the pathogenesis of Alzheimer’s disease (AD), a neurodegenerative condition in which an accumulation of intracellular neurofibrillary tangles and extracellular fibrillar amyloid beta is notably observed in the central nervous system. Genome-wide association studies have shown, both in microglia and astrocytes, an increase in gene variants associated with a higher risk of developing late-onset AD. Microglia, the resident innate immune cells of the brain, and astrocytes, glial cells crucial for vascular integrity and neuronal support, both agglomerate near amyloid beta plaques and dystrophic neurites where they participate in the elimination of these harmful parenchymal elements. However, their role in AD pathogenesis has been challenging to resolve due to the highly heterogeneous nature of these cell populations, i.e., their molecular, morphological, and ultrastructural diversity, together with their ever-changing responsiveness and functions throughout the pathological course of AD. With the recent expansions in the field of glial heterogeneity through innovative advances in state-of-the-art microscopy and -omics techniques, novel concepts and questions arose, notably pertaining to how the diverse microglial and astrocytic states interact with each other and with the AD hallmarks, and how their concerted efforts/actions impact the progression of the disease. In this review, we discuss the recent advances and findings on the topic of glial heterogeneity, particularly focusing on the relationships of these cells with AD hallmarks (e.g., amyloid beta plaques, neurofibrillary tangles, synaptic loss, and dystrophic neurites) in murine models of AD pathology and post-mortem brain samples of patients with AD.
Collapse
Affiliation(s)
- Marie-Kim St-Pierre
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jared VanderZwaag
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Sophia Loewen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Marie-Ève Tremblay
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
33
|
Zhu Y, Webster MJ, Murphy CE, Middleton FA, Massa PT, Liu C, Dai R, Weickert CS. Distinct Phenotypes of Inflammation Associated Macrophages and Microglia in the Prefrontal Cortex Schizophrenia Compared to Controls. Front Neurosci 2022; 16:858989. [PMID: 35844224 PMCID: PMC9279891 DOI: 10.3389/fnins.2022.858989] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/01/2022] [Indexed: 12/23/2022] Open
Abstract
Approximately 40% of people with schizophrenia are classified as having "high inflammation." This subgroup has worse neuropathology than patients with "low inflammation." Thus, one would expect the resident microglia and possibly monocyte-derived macrophages infiltrating from the periphery to be "activated" in those with schizophrenia with elevated neuroinflammation. To test whether microglia and/or macrophages are associated with increased inflammatory signaling in schizophrenia, we measured microglia- and macrophage-associated transcripts in the postmortem dorsolateral prefrontal cortex of 69 controls and 72 people with schizophrenia. Both groups were stratified by neuroinflammatory status based on cortical mRNA levels of cytokines and SERPINA3. We found microglial mRNAs levels were either unchanged (IBA1 and Hexb, p > 0.20) or decreased (CD11c, <62% p < 0.001) in high inflammation schizophrenia compared to controls. Conversely, macrophage CD163 mRNA levels were increased in patients, substantially so in the high inflammation schizophrenia subgroup compared to low inflammation subgroup (>250%, p < 0.0001). In contrast, high inflammation controls did not have elevated CD163 mRNA compared to low inflammation controls (p > 0.05). The pro-inflammatory macrophage marker (CD64 mRNA) was elevated (>160%, all p < 0.05) and more related to CD163 mRNA in the high inflammation schizophrenia subgroup compared to high inflammation controls, while anti-inflammatory macrophage and cytokine markers (CD206 and IL-10 mRNAs) were either unchanged or decreased in schizophrenia. Finally, macrophage recruitment chemokine CCL2 mRNA was increased in schizophrenia (>200%, p < 0.0001) and CCL2 mRNA levels positively correlated with CD163 mRNA (r = 0.46, p < 0.0001). Collectively, our findings support the co-existence of quiescent microglia and increased pro-inflammatory macrophages in the cortex of people with schizophrenia.
Collapse
Affiliation(s)
- Yunting Zhu
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Maree J. Webster
- Stanley Medical Research Institute, Rockville, MD, United States
| | - Caitlin E. Murphy
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
| | - Frank A. Middleton
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Paul T. Massa
- Department of Neurology and Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Cyndi Shannon Weickert
- Department of Neuroscience, SUNY Upstate Medical University, Syracuse, NY, United States
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
34
|
Jäntti H, Sitnikova V, Ishchenko Y, Shakirzyanova A, Giudice L, Ugidos IF, Gómez-Budia M, Korvenlaita N, Ohtonen S, Belaya I, Fazaludeen F, Mikhailov N, Gotkiewicz M, Ketola K, Lehtonen Š, Koistinaho J, Kanninen KM, Hernández D, Pébay A, Giugno R, Korhonen P, Giniatullin R, Malm T. Microglial amyloid beta clearance is driven by PIEZO1 channels. J Neuroinflammation 2022; 19:147. [PMID: 35706029 PMCID: PMC9199162 DOI: 10.1186/s12974-022-02486-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
Background Microglia are the endogenous immune cells of the brain and act as sensors of pathology to maintain brain homeostasis and eliminate potential threats. In Alzheimer's disease (AD), toxic amyloid beta (Aβ) accumulates in the brain and forms stiff plaques. In late-onset AD accounting for 95% of all cases, this is thought to be due to reduced clearance of Aβ. Human genome-wide association studies and animal models suggest that reduced clearance results from aberrant function of microglia. While the impact of neurochemical pathways on microglia had been broadly studied, mechanical receptors regulating microglial functions remain largely unexplored. Methods Here we showed that a mechanotransduction ion channel, PIEZO1, is expressed and functional in human and mouse microglia. We used a small molecule agonist, Yoda1, to study how activation of PIEZO1 affects AD-related functions in human induced pluripotent stem cell (iPSC)-derived microglia-like cells (iMGL) under controlled laboratory experiments. Cell survival, metabolism, phagocytosis and lysosomal activity were assessed using real-time functional assays. To evaluate the effect of activation of PIEZO1 in vivo, 5-month-old 5xFAD male mice were infused daily with Yoda1 for two weeks through intracranial cannulas. Microglial Iba1 expression and Aβ pathology were quantified with immunohistochemistry and confocal microscopy. Published human and mouse AD datasets were used for in-depth analysis of PIEZO1 gene expression and related pathways in microglial subpopulations. Results We show that PIEZO1 orchestrates Aβ clearance by enhancing microglial survival, phagocytosis, and lysosomal activity. Aβ inhibited PIEZO1-mediated calcium transients, whereas activation of PIEZO1 with a selective agonist, Yoda1, improved microglial phagocytosis resulting in Aβ clearance both in human and mouse models of AD. Moreover, PIEZO1 expression was associated with a unique microglial transcriptional phenotype in AD as indicated by assessment of cellular metabolism, and human and mouse single-cell datasets. Conclusion These results indicate that the compromised function of microglia in AD could be improved by controlled activation of PIEZO1 channels resulting in alleviated Aβ burden. Pharmacological regulation of these mechanoreceptors in microglia could represent a novel therapeutic paradigm for AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02486-y.
Collapse
Affiliation(s)
- Henna Jäntti
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Valeriia Sitnikova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Yevheniia Ishchenko
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.,Departments of Molecular Biophysics and Biochemistry and Neuroscience, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Anastasia Shakirzyanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Luca Giudice
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.,Department of Computer Science, University of Verona, 37134, Verona, Italy
| | - Irene F Ugidos
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Mireia Gómez-Budia
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Nea Korvenlaita
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Sohvi Ohtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Irina Belaya
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Feroze Fazaludeen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Nikita Mikhailov
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Maria Gotkiewicz
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Kirsi Ketola
- Institute of Biomedicine, University of Eastern Finland, 70210, Kuopio, Finland
| | - Šárka Lehtonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.,Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Katja M Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Damian Hernández
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, 37134, Verona, Italy
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.
| |
Collapse
|
35
|
Kiani Shabestari S, Morabito S, Danhash EP, McQuade A, Sanchez JR, Miyoshi E, Chadarevian JP, Claes C, Coburn MA, Hasselmann J, Hidalgo J, Tran KN, Martini AC, Chang Rothermich W, Pascual J, Head E, Hume DA, Pridans C, Davtyan H, Swarup V, Blurton-Jones M. Absence of microglia promotes diverse pathologies and early lethality in Alzheimer's disease mice. Cell Rep 2022; 39:110961. [PMID: 35705056 PMCID: PMC9285116 DOI: 10.1016/j.celrep.2022.110961] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 04/13/2022] [Accepted: 05/24/2022] [Indexed: 11/03/2022] Open
Abstract
Microglia are strongly implicated in the development and progression of Alzheimer's disease (AD), yet their impact on pathology and lifespan remains unclear. Here we utilize a CSF1R hypomorphic mouse to generate a model of AD that genetically lacks microglia. The resulting microglial-deficient mice exhibit a profound shift from parenchymal amyloid plaques to cerebral amyloid angiopathy (CAA), which is accompanied by numerous transcriptional changes, greatly increased brain calcification and hemorrhages, and premature lethality. Remarkably, a single injection of wild-type microglia into adult mice repopulates the microglial niche and prevents each of these pathological changes. Taken together, these results indicate the protective functions of microglia in reducing CAA, blood-brain barrier dysfunction, and brain calcification. To further understand the clinical implications of these findings, human AD tissue and iPSC-microglia were examined, providing evidence that microglia phagocytose calcium crystals, and this process is impaired by loss of the AD risk gene, TREM2.
Collapse
Affiliation(s)
- Sepideh Kiani Shabestari
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Samuel Morabito
- Mathematical, Computational and System Biology (MCSB) Program, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Emma Pascal Danhash
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Amanda McQuade
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Jessica Ramirez Sanchez
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Emily Miyoshi
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA
| | - Jean Paul Chadarevian
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Christel Claes
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Morgan Alexandra Coburn
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Jonathan Hasselmann
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Jorge Hidalgo
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Kayla Nhi Tran
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA; Department of Pathology & Laboratory Medicine, UC Irvine, Irvine, CA 92697, USA
| | | | - Jesse Pascual
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA; Department of Pathology & Laboratory Medicine, UC Irvine, Irvine, CA 92697, USA
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA; Department of Pathology & Laboratory Medicine, UC Irvine, Irvine, CA 92697, USA
| | - David A Hume
- Mater Research Institute-University of Queensland, Brisbane, Australia
| | - Clare Pridans
- University of Edinburgh Centre for Inflammation Research, Edinburgh, UK; Simons Initiative for the Developing Brain Centre, University of Edinburgh, Edinburgh, UK; The Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
| | - Hayk Davtyan
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Vivek Swarup
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
36
|
Pharmacological Activation of GPR55 Improved Cognitive Impairment Induced by Lipopolysaccharide in Mice. J Mol Neurosci 2022; 72:1656-1669. [PMID: 35596056 DOI: 10.1007/s12031-022-02020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
Our previous research found that activation of GPR55 can alleviate cognitive impairment induced by amyloid-beta 1-42 (Aβ1-42) and streptozotocin in mice, but the role of GPR55 in the pathogenesis of cognitive impairment remains unknown. Here, we used a lipopolysaccharide (LPS) mouse model to further investigate the role and mechanism of O-1602, a GPR55 agonist, on cognitive dysfunction. ICR mice were treated with an intracerebroventricular (i.c.v.) injection of LPS, followed by cognitive function tests. The expression of GPR55, NF-κB p65, caspase-3, Bax, and Bcl-2 in the hippocampus was examined by Western blotting. Inflammatory cytokines and microglia were detected by ELISA kit and immunohistochemical analyses, respectively. The levels of MDA, GSH, SOD, and CAT were examined by assay kits. Furthermore, TUNEL-staining was used to detect neuronal apoptosis. Our results showed that i.c.v. injection of LPS in mice exhibited impaired performance in the behavior tests, which were ameliorated by O-1602 treatment (2.0 or 4.0 μg/mouse, i.c.v.). Importantly, we found that O-1602 treatment reversed GPR55 downregulation, decreased the expression of NF-κB p65, suppressed the accumulation of proinflammatory cytokines and microglia activation, increased the anti-inflammatory cytokines, and reduced the levels of MDA, increased the levels of GSH, SOD, and CAT in the hippocampus. In addition, O-1602 treatment also significantly reduced Bax and increased Bcl-2 expression as well as decreased caspase-3 activity and TUNEL-positive cells in the hippocampus. These observations indicate that O-1602 may ameliorate LPS-induced cognition deficits via inhibiting neuroinflammation, oxidative stress, and apoptosis mediated by the NF-κB pathway in mice.
Collapse
|
37
|
Zhou R, Qian S, Cho WCS, Zhou J, Jin C, Zhong Y, Wang J, Zhang X, Xu Z, Tian M, Chan LWC, Zhang H. Microbiota-microglia connections in age-related cognition decline. Aging Cell 2022; 21:e13599. [PMID: 35349746 PMCID: PMC9124309 DOI: 10.1111/acel.13599] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable process that all individuals experience, of which the extent differs among individuals. It has been recognized as the risk factor of neurodegenerative diseases by affecting gut microbiota compositions, microglia, and cognition abilities. Aging‐induced changes in gut microbiota compositions have a critical role in orchestrating the morphology and functions of microglia through the gut‐brain axis. Gut microbiota communicates with microglia by its secreted metabolites and neurotransmitters. This is highly associated with age‐related cognitive declines. Here, we review the main composition of microbiota in the aged individuals, outline the changes of the brain in age‐related cognitive decline from a neuroinflammation perspective, especially the changes of morphology and functions of microglia, discuss the crosstalk between microbiota and microglia in the aged brain and further highlight the role of microbiota‐microglia connections in neurodegenerative diseases (Alzheimer's disease and Parkinson's disease).
Collapse
Affiliation(s)
- Rui Zhou
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Hong Kong SAR China
| | - Shufang Qian
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - William C. S. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Hong Kong SAR China
| | - Jinyun Zhou
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Chentao Jin
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Yan Zhong
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Jing Wang
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Xiaohui Zhang
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Zhoujiao Xu
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Mei Tian
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - Lawrence W. C. Chan
- Department of Health Technology and Informatics The Hong Kong Polytechnic University Hong Kong SAR China
| | - Hong Zhang
- Department of Nuclear Medicine and Medical PET Center The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
- Key Laboratory for Biomedical Engineering of Ministry of Education Zhejiang University Hangzhou China
- The College of Biomedical Engineering and Instrument Science of Zhejiang University Hangzhou China
| |
Collapse
|
38
|
Patten KT, Valenzuela AE, Wallis C, Harvey DJ, Bein KJ, Wexler AS, Gorin FA, Lein PJ. Hippocampal but Not Serum Cytokine Levels Are Altered by Traffic-Related Air Pollution in TgF344-AD and Wildtype Fischer 344 Rats in a Sex- and Age-Dependent Manner. Front Cell Neurosci 2022; 16:861733. [PMID: 35530180 PMCID: PMC9072828 DOI: 10.3389/fncel.2022.861733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/24/2022] [Indexed: 11/19/2022] Open
Abstract
Epidemiological studies have demonstrated that air pollution is a significant risk factor for age-related dementia, including Alzheimer's disease (AD). It has been posited that traffic-related air pollution (TRAP) promotes AD neuropathology by exacerbating neuroinflammation. To test this hypothesis, serum and hippocampal cytokines were quantified in male and female TgF344-AD rats and wildtype (WT) Fischer 344 littermates exposed to TRAP or filtered air (FA) from 1 to 15 months of age. Luminex™ rat 23-cytokine panel assays were used to measure the levels of hippocampal and serum cytokines in 3-, 6-, 10-, and 15-month-old rats (corresponding to 2, 5, 9, and 14 months of exposure, respectively). Age had a pronounced effect on both serum and hippocampal cytokines; however, age-related changes in hippocampus were not mirrored in the serum and vice versa. Age-related changes in serum cytokine levels were not influenced by sex, genotype, or TRAP exposure. However, in the hippocampus, in 3-month-old TgF344-AD and WT animals, TRAP increased IL-1ß in females while increasing TNF ɑin males. In 6-month-old animals, TRAP increased hippocampal levels of M-CSF in TgF344-AD and WT females but had no significant effect in males. At 10 and 15 months of age, there were minimal effects of TRAP, genotype or sex on hippocampal cytokines. These observations demonstrate that TRAP triggers an early inflammatory response in the hippocampus that differs with sex and age and is not reflected in the serum cytokine profile. The relationship of TRAP effects on cytokines to disease progression remains to be determined.
Collapse
Affiliation(s)
- Kelley T. Patten
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Anthony E. Valenzuela
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Christopher Wallis
- Air Quality Research Center, University of California, Davis, Davis, CA, United States
| | - Danielle J. Harvey
- Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Keith J. Bein
- Air Quality Research Center, University of California, Davis, Davis, CA, United States
- Center for Health and the Environment, University of California, Davis, Davis, CA, United States
| | - Anthony S. Wexler
- Air Quality Research Center, University of California, Davis, Davis, CA, United States
- Mechanical and Aerospace Engineering, Civil and Environmental Engineering, College of Engineering, University of California, Davis, Davis, CA, United States
- Land, Air and Water Resources, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA, United States
| | - Fredric A. Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Department of Neurology, Davis School of Medicine, University of California, Sacramento, Sacramento, CA, United States
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
39
|
Lynch MA. Exploring Sex-Related Differences in Microglia May Be a Game-Changer in Precision Medicine. Front Aging Neurosci 2022; 14:868448. [PMID: 35431903 PMCID: PMC9009390 DOI: 10.3389/fnagi.2022.868448] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/08/2022] [Indexed: 12/16/2022] Open
Abstract
One area of microglial biology that has been relatively neglected until recently is sex differences and this is in spite of the fact that sex is a risk factor in several diseases that are characterized by neuroinflammation and, by extension, microglial activation. Why these sex differences exist is not known but the panoply of differences extend to microglial number, genotype and phenotype. Significantly, several of these sex-related differences are also evident in health and change during life emphasizing the dynamic and plastic nature of microglia. This review will consider how age impacts on sex-related differences in microglia and ask whether the advancement of personalized medicine demands that a greater focus is placed on studying sex-related differences in microglia in Alzheimer's disease, Parkinson's disease and models of inflammatory stress and trauma in order to make true progress in dealing with these conditions.
Collapse
Affiliation(s)
- Marina A. Lynch
- Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
40
|
Colony-stimulating factor 1 receptor signaling in the central nervous system and the potential of its pharmacological inhibitors to halt the progression of neurological disorders. Inflammopharmacology 2022; 30:821-842. [PMID: 35290551 DOI: 10.1007/s10787-022-00958-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Colony Stimulating Factor-1 (CSF-1)/Colony Stimulating Factor-1 Receptor (CSF-1R) signaling axis plays an essential role in the development, maintenance, and proliferation of macrophage lineage cells. Within the central nervous system, CSF-1R signaling primarily maintains microglial homeostasis. Microglia, being the resident macrophage and first responder to any neurological insults, plays critical importance in overall health of the human brain. Aberrant and sustained activation of microglia along with continued proliferation and release of neurotoxic proinflammatory cytokines have been reported in various neurological and neurodegenerative diseases. Therefore, halting the neuroinflammatory pathway via targeting microglial proliferation, which depends on CSF-1R signaling, has emerged as a potential therapeutic target for neurological disorders. However, apart from regulating the microglial function, recently it has been discovered that CSF-1R has much broader role in central nervous system. These findings limit the therapeutic utility of CSF-1R inhibitors but also highlight the need for a complete understanding of CSF-1R function within the central nervous system. Moreover, it has been found that selective inhibitors of CSF-1R may be more efficient in avoiding non-specific targeting and associated side effects. Short-term depletion of microglial population in diseased conditions have also been found to be beneficial; however, the dose and therapeutic window for optimum effects may need to be standardized further.This review summarizes the present understanding of CSF-1R function within the central nervous system. We discuss the CSF-1R signaling in the context of microglia function, crosstalk between microglia and astroglia, and regulation of neuronal cell function. We also discuss a few of the neurological disorders with a focus on the utility of CSF-1R inhibitors as potential therapeutic strategy for halting the progression of neurological diseases.
Collapse
|
41
|
Li Q, Wu Y, Chen J, Xuan A, Wang X. Microglia and immunotherapy in Alzheimer's disease. Acta Neurol Scand 2022; 145:273-278. [PMID: 34779509 DOI: 10.1111/ane.13551] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/21/2021] [Accepted: 10/31/2021] [Indexed: 01/01/2023]
Abstract
Microglia are a type of glial cells that play a key role in the repair of damage to the central nervous system (CNS). In the pathological condition of Alzheimer's disease (AD), β-amyloid peptide and a variety of pro-inflammatory factors can activate microglia, resulting in the secretion of a variety of inflammatory factors and neurotoxins. This leads to neuronal damage and even apoptosis, thus triggering AD. In contrast, microglia can protect the CNS by phagocytizing Aβ to slow down AD development. In this review, the roles of microglia in AD neuroinflammation and the scope of immunotherapy for AD are summarized to provide a theoretical basis for AD prevention and treatment.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Yingying Wu
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Jichun Chen
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Aiguo Xuan
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Xiao Wang
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| |
Collapse
|
42
|
Gao H, Ju F, Ti R, Zhang Y, Zhang S. Differential Regulation of Microglial Activation in Response to Different Degree of Ischemia. Front Immunol 2022; 13:792638. [PMID: 35154109 PMCID: PMC8831277 DOI: 10.3389/fimmu.2022.792638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are primary immune cells within the brain and are rapidly activated after cerebral ischemia. The degree of microglial activation is closely associated with the severity of ischemia. However, it remains largely unclear how microglial activation is differentially regulated in response to a different degree of ischemia. In this study, we used a bilateral common carotid artery ligation (BCAL) model and induced different degrees of ischemia by varying the duration of ligation to investigate the microglial response in CX3CR1GFP/+ mice. Confocal microscopy, immunofluorescence staining, RNA sequencing, and qRT-PCR were used to evaluate the de-ramification, proliferation, and differential gene expression associated with microglial activation. Our results showed that 30 min of ischemia induced rapid de-ramification of microglia but did not have significant influence on the microglial density. In contrast, 60 min of ischemia led to a significant decrease in microglial density and more pronounced de-ramification of microglial processes. Importantly, 30 min of ischemia did not induce proliferation of microglia, but 60 min of ischemia led to a marked increase in the density of proliferative microglia. Further analysis utilized transcriptome sequencing showed that microglial activation is differentially regulated in response to different degrees of ischemia. A total of 1,097 genes were differentially regulated after 60 min of ischemia, but only 68 genes were differentially regulated after 30 min of ischemia. Pathway enrichment analysis showed that apoptosis, cell mitosis, immune receptor activity and inflammatory-related pathways were highly regulated after 60 min of ischemia compared to 30 min of ischemia. Multiple microglia-related genes such as Cxcl10, Tlr7, Cd86, Tnfrsf1a, Nfkbia, Tgfb1, Ccl2 and Il-6, were upregulated with prolonged ischemia. Pharmacological inhibition of CSF1 receptor demonstrated that CSF1R signaling pathway contributed to microglial proliferation. Together, these results suggest that the proliferation of microglia is gated by the duration of ischemia and microglia were differentially activated in responding to different degrees of ischemia.
Collapse
|
43
|
Neuroimmune contributions to Alzheimer's disease: a focus on human data. Mol Psychiatry 2022; 27:3164-3181. [PMID: 35668160 PMCID: PMC9168642 DOI: 10.1038/s41380-022-01637-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
The past decade has seen the convergence of a series of new insights that arose from genetic and systems analyses of Alzheimer's disease (AD) with a wealth of epidemiological data from a variety of fields; this resulted in renewed interest in immune responses as important, potentially causal components of AD. Here, we focus primarily on a review of human data which has recently yielded a set of robust, reproducible results that exist in a much larger universe of conflicting reports stemming from small studies with important limitations in their study design. Thus, we are at an important crossroads in efforts to first understand at which step of the long, multiphasic course of AD a given immune response may play a causal role and then modulate this response to slow or block the pathophysiology of AD. We have a wealth of new experimental tools, analysis methods, and capacity to sample human participants at large scale longitudinally; these resources, when coupled to a foundation of reproducible results and novel study designs, will enable us to monitor human immune function in the CNS at the level of complexity that is required while simultaneously capturing the state of the peripheral immune system. This integration of peripheral and central perturbations in immune responses results in pathologic responses in the central nervous system parenchyma where specialized cellular microenvironments composed of multiple cell subtypes respond to these immune perturbations as well as to environmental exposures, comorbidities and the impact of the advancing life course. Here, we offer an overview that seeks to illustrate the large number of interconnecting factors that ultimately yield the neuroimmune component of AD.
Collapse
|
44
|
Mou Q, Yao K, Ye M, Zhao B, Hu Y, Lou X, Li H, Zhang H, Zhao Y. Modulation of Sirt1-mTORC1 Pathway in Microglia Attenuates Retinal Ganglion Cell Loss After Optic Nerve Injury. J Inflamm Res 2021; 14:6857-6869. [PMID: 34934336 PMCID: PMC8684404 DOI: 10.2147/jir.s338815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose Optic nerve injury (ONI) causes neuroinflammation and neurodegeneration leading to visual deficits. The response of microglia has emerged as an impactful component of etiology in neurodegeneration. This study aimed to investigate the effect of SIRT1-mTORC1 signaling pathway in microglia regulation after ONI. Methods Cx3Cr1-CreERT2/RaptorF/F and Cx3Cr1-CreERT2/Sirt1F/F mice were used to delete Raptor and Sirt1 in microglia, respectively. Optic nerve crush (ONC) model was established to mimic ONI. PLX5622, a highly specific inhibitor of the colony-stimulating factor 1 receptor (CSF1R), is used to eliminate microglia in optic nerve. Ionized calcium binding adaptor molecule 1 (Iba1) immunostaining was used to detect microglial activation. Retinal ganglion cells (RGCs) were quantified by Nissl staining and retinal whole-mount immunostaining with RNA-binding protein with multiple splicing (RBPMS). Axonal damage was valued by transmission electron microscopy (TEM). Results Microglial activation emerged on day 3 post ONC and was earlier than RGCs loss which occurred at day 5 after injury. Depleting microglia with PLX5622 could attenuate the loss of RGCs and axon damage after ONC. Gain- and loss-of-function studies revealed that SIRT1 determined the activation of microglia in optic nerve. In addition, microglia-specific deletion of Raptor resulted in decreased microglial activation. Interestingly, activating mTORC1 with CCT007093 could reverse the function of SIRT1 in regulating the process of microglial activation mediated RGCs loss. Conclusion Our study reveals a potential novel mechanism of SIRT1-mTORC1 pathway in microglia regulation, and indicates a therapeutic potential for the protection of RGCs in ONI.
Collapse
Affiliation(s)
- Qianxue Mou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ke Yao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Meng Ye
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yuanyuan Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiaotong Lou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Huixia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| |
Collapse
|
45
|
Pan J, Ma N, Zhong J, Yu B, Wan J, Zhang W. Age-associated changes in microglia and astrocytes ameliorate blood-brain barrier dysfunction. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:970-986. [PMID: 34760339 PMCID: PMC8561003 DOI: 10.1016/j.omtn.2021.08.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) dysfunction is associated with an accumulation of neurotoxic molecules and increased infiltration of peripheral cells within the brain parenchyma. Accruing evidence suggests that microglia and astrocytes play a crucial role in the recovery of BBB integrity and the corralling of infiltrating cells into clusters after brain damage, but the mechanisms involved remain unclear. Intriguingly, the results of flow cytometry and immunofluorescence analyses have shown that BBB permeability to peripheral cells is substantially enhanced during normal aging at 12 months in mice. Thus, we used the SMART-seq2 method to perform RNA sequencing of microglia and astrocytes at five time points before and immediately after the BBB permeability change. Our comprehensive analyses revealed that microglia are characterized by marked alterations in the negative regulation of protein phosphorylation and phagocytic vesicles, whereas astrocytes show elevated enzyme or peptidase-inhibitor activity in the recovery of BBB function. Moreover, we identified a cassette of key genes that might ameliorate the insults of pathophysiological events in aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Jie Pan
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China.,Department of Pathology and Neuropathology, Stanford University School of Medicine, CA 94305, USA
| | - Nana Ma
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| | - Jie Zhong
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China
| | - Bo Yu
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province, China.,Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Jun Wan
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China.,Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong Province, PRC
| | - Wei Zhang
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong Province, China
| |
Collapse
|
46
|
Lin JZ, Duan MR, Lin N, Zhao WJ. The emerging role of the chondroitin sulfate proteoglycan family in neurodegenerative diseases. Rev Neurosci 2021; 32:737-750. [PMID: 33655733 DOI: 10.1515/revneuro-2020-0146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/07/2021] [Indexed: 02/05/2023]
Abstract
Chondroitin sulfate (CS) is a kind of linear polysaccharide that is covalently linked to proteins to form proteoglycans. Chondroitin sulfate proteoglycans (CSPGs) consist of a core protein, with one or more CS chains covalently attached. CSPGs are precisely regulated and they exert a variety of physiological functions by binding to adhesion molecules and growth factors. Widely distributed in the nervous system in human body, CSPGs contribute to the major component of extracellular matrix (ECM), where they play an important role in the development and maturation of the nervous system, as well as in the pathophysiological response to damage to the central nervous system (CNS). While there are more than 30 types of CSPGs, this review covers the roles of the most important ones, including versican, aggrecan, neurocan and NG2 in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. The updated reports of the treatment of neurodegenerative diseases are involving CSPGs.
Collapse
Affiliation(s)
- Jia-Zhe Lin
- Neurosurgical Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Ming-Rui Duan
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Nuan Lin
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
47
|
Comparative Review of Microglia and Monocytes in CNS Phagocytosis. Cells 2021; 10:cells10102555. [PMID: 34685535 PMCID: PMC8534258 DOI: 10.3390/cells10102555] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023] Open
Abstract
Macrophages maintain tissue homeostasis by phagocytosing and removing unwanted materials such as dead cells and cell debris. Microglia, the resident macrophages of the central nervous system (CNS), are no exception. In addition, a series of recent studies have shown that microglia phagocytose the neuronal synapses that form the basis of neural circuit function. This discovery has spurred many neuroscientists to study microglia. Importantly, in the CNS parenchyma, not only microglia but also blood-derived monocytes, which essentially differentiate into macrophages after infiltration, exert phagocytic ability, making the study of phagocytosis in the CNS even more interesting and complex. In particular, in the diseased brain, the phagocytosis of tissue-damaging substances, such as myelin debris in multiple sclerosis (MS), has been shown to be carried out by both microglia and blood-derived monocytes. However, it remains largely unclear why blood-derived monocytes need to invade the parenchyma, where microglia are already abundant, to assist in phagocytosis. We will also discuss whether this phagocytosis can affect the fate of the phagocytosing cell itself as well as the substance being phagocytosed and the surrounding environment in addition to future research directions. In this review, we will introduce recent studies to answer a question that often arises when studying microglial phagocytosis: under what circumstances and to what extent blood-derived monocytes infiltrate the CNS and contribute to phagocytosis. In addition, the readers will learn how recent studies have experimentally distinguished between microglia and infiltrating monocytes. Finally, we aim to contribute to the progress of phagocytosis research by discussing the effects of phagocytosis on phagocytic cells.
Collapse
|
48
|
Qiu H, Zhao R, Fei G, Pan X, Sang S, Xu Y, Jin B, Jin L, Cheng X, Zhong C. Dynamic Change of Intracellular Metabolism of Microglia Evaluated by Transcriptomics in an Alzheimer's Mouse Model. J Alzheimers Dis 2021; 81:517-531. [PMID: 33814454 DOI: 10.3233/jad-210213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Microglia play diverse roles in Alzheimer's disease (AD). Intracellular metabolism has been indicated an important factor in modulating the function of microglia. However, it is not clear whether the intracellular metabolism of microglia changes dynamically in different stages of AD. OBJECTIVE To determine whether microglia intracellular metabolism changes dynamically in different stages of AD. METHODS Microglia were extracted from APPSwe/PS1dE9 (APP/PS1) mice and wild-type littermates at 2, 4, and 8 months old by fluorescence-activated cell sorting and used for RNA-sequencing analysis and quantitative PCR. Morphologies of amyloid plaques and microglia were detected by immunofluorescence staining. RESULTS Compared with control littermates, the microglia of APP/PS1 mice exhibited significant transcriptional changes at 2-month-old before microglia morphological alterations and the plaque formation. The changes continued drastically following age with defined morphological shift of microglia and amyloid plaque enhancement in brains. Further analysis of those genotype and age dependent transcriptomic changes revealed that differentially expressed genes were enriched in pathways related to energy metabolism. Compared with wild-type mice, there were changes of some vital genes related to glucose metabolism and lipid metabolism pathways in APP/PS1 mice at different ages. Glucose metabolism may play a major role in early activation of microglia, and lipid metabolism may be more important in later activation period. CONCLUSION Our results showed that microglia actively participate in the pathological progress of AD. The intracellular metabolism of microglia changed significantly in different stages of AD, even preceding amyloid-β deposition.
Collapse
Affiliation(s)
- Hongyan Qiu
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ruoqi Zhao
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Guoqiang Fei
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.,Department of Neurology, Xiamen Branch Zhongshan Hospital, Fudan University, Xiamen, China
| | - Xiaoli Pan
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Shaoming Sang
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yangqi Xu
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Boru Jin
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lirong Jin
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Abcouwer SF, Shanmugam S, Muthusamy A, Lin CM, Kong D, Hager H, Liu X, Antonetti DA. Inflammatory resolution and vascular barrier restoration after retinal ischemia reperfusion injury. J Neuroinflammation 2021; 18:186. [PMID: 34446062 PMCID: PMC8394696 DOI: 10.1186/s12974-021-02237-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023] Open
Abstract
Background Several retinal pathologies exhibit both inflammation and breakdown of the inner blood-retinal barrier (iBRB) resulting in vascular permeability, suggesting that treatments that trigger resolution of inflammation may also promote iBRB restoration. Methods Using the mouse retinal ischemia-reperfusion (IR) injury model, we followed the time course of neurodegeneration, inflammation, and iBRB disruption and repair to examine the relationship between resolution of inflammation and iBRB restoration and to determine if minocycline, a tetracycline derivative shown to reverse microglial activation, can hasten these processes. Results A 90-min ischemic insult followed by reperfusion in the retina induced cell apoptosis and inner retina thinning that progressed for approximately 2 weeks. IR increased vascular permeability within hours, which resolved between 3 and 4 weeks after injury. Increased vascular permeability coincided with alteration and loss of endothelial cell tight junction (TJ) protein content and disorganization of TJ protein complexes. Shunting of blood flow away from leaky vessels and dropout of leaky capillaries were eliminated as possible mechanisms for restoring the iBRB. Repletion of TJ protein contents occurred within 2 days after injury, long before restoration of the iBRB. In contrast, the eventual re-organization of TJ complexes at the cell border coincided with restoration of the barrier. A robust inflammatory response was evident a 1 day after IR and progressed to resolution over the 4-week time course. The inflammatory response included a rapid and transient infiltration of granulocytes and Ly6C+ classical inflammatory monocytes, a slow accumulation of Ly6Cneg monocyte/macrophages, and activation, proliferation, and mobilization of resident microglia. Extravasation of the majority of CD45+ leukocytes occurred from the superficial plexus. The presence of monocyte/macrophages and increased numbers of microglia were sustained until the iBRB was eventually restored. Intervention with minocycline to reverse microglial activation at 1 week after injury promoted early restoration of the iBRB coinciding with decreased expression of mRNAs for the microglial M1 markers TNF-α, IL-1β, and Ptgs2 (Cox-2) and increased expression of secreted serine protease inhibitor Serpina3n mRNA. Conclusions These results suggest that iBRB restoration occurs as TJ complexes are reorganized and that resolution of inflammation and restoration of the iBRB following retinal IR injury are functionally linked. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02237-5.
Collapse
Affiliation(s)
- Steven F Abcouwer
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA.
| | - Sumathi Shanmugam
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | | | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Dejuan Kong
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Xuwen Liu
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, Michigan Medicine, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, 48105, USA.,Department of Molecular and Integrative Physiology, Ann Arbor, MI, 48109, USA
| |
Collapse
|
50
|
Mehina EMF, Taylor S, Boghozian R, White E, Choi SE, Cheema MS, Korbelin J, Brown CE. Invasion of phagocytic Galectin 3 expressing macrophages in the diabetic brain disrupts vascular repair. SCIENCE ADVANCES 2021; 7:7/34/eabg2712. [PMID: 34407943 PMCID: PMC8373127 DOI: 10.1126/sciadv.abg2712] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/28/2021] [Indexed: 06/13/2023]
Abstract
The cellular events that dictate the repair of damaged vessels in the brain, especially in those with vascular risk factors such as diabetes, is poorly understood. Here, we dissected the role of resident microglia and infiltrative macrophages in determining the repair of ruptured cerebral microvessels. Using in vivo time-lapse imaging, gene expression analysis, and immunohistochemistry, we identified a unique population of phagocytic Galectin 3 (Gal3) expressing macrophages, distinct from resident microglia, which infiltrated and aggregated at the site of injury in diabetic mice and were associated with the elimination of microvessels. Depletion of these infiltrative macrophages in diabetic mice attenuated phagocytic activity and prevented the loss of blood vessels after injury. These findings highlight a previously unknown role for infiltrative Gal3 expressing macrophages in promoting vessel elimination after brain injury and provide impetus for future studies to determine whether depleting these cells can facilitate vascular repair in at risk populations.
Collapse
Affiliation(s)
- Eslam M F Mehina
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Stephanie Taylor
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Roobina Boghozian
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Emily White
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Sun Eui Choi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Manjinder S Cheema
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jakob Korbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|