1
|
Wang M, Li T, Li W, Song T, Zhao C, Wu Q, Cui W, Hao Y, Hou Y, Zhu P. Unraveling the neuroprotective potential of scalp electroacupuncture in ischemic stroke: A key role for electrical stimulation. Neuroscience 2024; 562:160-181. [PMID: 39401739 DOI: 10.1016/j.neuroscience.2024.10.019] [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: 08/21/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
This study aims to explore the neuroprotective effects of scalp Electroacupuncture (EA) on ischemic stroke, with a specific focus on the role of electrical stimulation (ES). Employing a rat model of middle cerebral artery occlusion (MCAO), we used methods such as Triphenyl tetrazolium chloride staining, micro-CT scanning, Enzyme linked immunosorbent assay (ELISA), and immunofluorescence to assess the impacts of EA. We further conducted RNA-seq analysis and in vitro experiments with organotypic brain slices and cerebral organoids to explore the underlying mechanisms. Our research revealed that EA notably reduced cerebral infarct volume and improved regional cerebral blood flow in rats following MCAO. Micro-CT imaging showed improved vascular integrity in EA-treated groups. Histological analyses, including HE staining, indicated reduced brain tissue damage. ELISA demonstrated a decrease in pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, suggesting improved blood-brain barrier function. Immunofluorescence and Western blot analyses revealed that EA treatment significantly inhibited microglial and astrocytic overactivation. RNA-seq analysis of brain tissues highlighted a downregulation of immune pathways and inflammatory responses, confirming the neuroprotective role of EA. This was further corroborated by in vitro experiments using organotypic brain slices and cerebral organoids, which showcased the efficacy of electrical stimulation in reducing neuroinflammation and protecting neuronal cells. The study highlights the potential of scalp EA, particularly its ES component, in treating ischemic stroke. It provides new insights into the mechanisms of EA, emphasizing its efficacy in neuroprotection and modulation of neuroinflammation, and suggests avenues for optimized treatment strategies in stroke therapy.
Collapse
Affiliation(s)
- Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tongtong Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Wenyan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tao Song
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Chi Zhao
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Qiulan Wu
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Wenwen Cui
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; National Key Laboratory for Innovation and Transformation of Luobing Theory, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Key Laboratory of State Administration of TCM Cardio-Cerebral Vessel Collateral Disease, No.238, the South of Tianshan Street, Shijiazhuang, 050035, Hebei, China.
| | - Pengyu Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 411, The Street of Guogeli, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
2
|
Peethambaran Mallika A. Tri-culture modeling of neuroinflammation, neurodegeneration, and neuroprotection. J Alzheimers Dis 2024; 102:742-744. [PMID: 39506313 DOI: 10.1177/13872877241294181] [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] [Indexed: 11/08/2024]
Abstract
The study of neurodegenerative diseases, such as Alzheimer's disease (AD), has long been a complex and challenging task. One of the major hurdles in understanding these diseases is the difficulty in recapitulating the complex interactions between neurons, astrocytes, and microglia in a laboratory setting. In recent years, researchers have made significant progress in developing triculture models that combine these three cell types, allowing for a more accurate representation of the cellular context of the human brain. This commentary discusses the recent advancements and importance of using tri-culture model systems in clarifying the pathophysiology of AD and discusses the recent article by Kim et al. (2024) published in the Journal of Alzheimer's Disease.
Collapse
|
3
|
Garcia DW, Jacquir S. Astrocyte-mediated neuronal irregularities and dynamics: the complexity of the tripartite synapse. BIOLOGICAL CYBERNETICS 2024; 118:249-266. [PMID: 39276225 DOI: 10.1007/s00422-024-00994-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/08/2024] [Indexed: 09/16/2024]
Abstract
Despite significant advancements in recent decades, gaining a comprehensive understanding of brain computations remains a significant challenge in neuroscience. Using computational models is crucial for unraveling this complex phenomenon and is equally indispensable for studying neurological disorders. This endeavor has created many neuronal models that capture brain dynamics at various scales and complexities. However, most existing models do not account for the potential influence of glial cells, particularly astrocytes, on neuronal physiology. This gap persists even with the emerging evidence indicating their critical role in regulating neural network activity, plasticity, and even neurological pathologies. To address this gap, some works proposed models that include neuron-glia interactions. Also, while some literature focuses on sophisticated models of neuron-glia interactions that mimic the complexity of physiological phenomena, there are also existing works that propose simplified models of neural-glial ensembles. Building upon these efforts, we aimed to contribute further to the field by proposing a simplified tripartite synapse model that encompasses the presynaptic neuron, postsynaptic neuron, and astrocyte. We defined the tripartite synapse model based on the Adaptive Exponential Integrate-and-Fire neuron model and a simplified scheme of the astrocyte model previously proposed by Postnov. Through our simulations, we demonstrated how astrocytes can influence neuronal firing behavior by sequentially activating and deactivating different pathways within the tripartite synapse. This modulation by astrocytes can shape neuronal behavior and introduce irregularities in the firing patterns of both presynaptic and postsynaptic neurons through the introduction of new pathways and configurations of relevant parameters.
Collapse
Affiliation(s)
- Den Whilrex Garcia
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, 91400, France.
- Department of Engineering, Lyceum of the Philippines University, Cavite, Philippines.
| | - Sabir Jacquir
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay, 91400, France.
| |
Collapse
|
4
|
Sabogal-Guaqueta AM, Mitchell-Garcia T, Hunneman J, Voshart D, Thiruvalluvan A, Foijer F, Kruyt F, Trombetta-Lima M, Eggen BJL, Boddeke E, Barazzuol L, Dolga AM. Brain organoid models for studying the function of iPSC-derived microglia in neurodegeneration and brain tumours. Neurobiol Dis 2024; 203:106742. [PMID: 39581553 DOI: 10.1016/j.nbd.2024.106742] [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: 09/30/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Microglia represent the main resident immune cells of the brain. The interplay between microglia and other cells in the central nervous system, such as neurons or other glial cells, influences the function and ability of microglia to respond to various stimuli. These cellular communications, when disrupted, can affect the structure and function of the brain, and the initiation and progression of neurodegenerative diseases including Alzheimer's disease and Parkinson's disease, as well as the progression of other brain diseases like glioblastoma. Due to the difficult access to patient brain tissue and the differences reported in the murine models, the available models to study the role of microglia in disease progression are limited. Pluripotent stem cell technology has facilitated the generation of highly complex models, allowing the study of control and patient-derived microglia in vitro. Moreover, the ability to generate brain organoids that can mimic the 3D tissue environment and intercellular interactions in the brain provide powerful tools to study cellular pathways under homeostatic conditions and various disease pathologies. In this review, we summarise the most recent developments in modelling degenerative diseases and glioblastoma, with a focus on brain organoids with integrated microglia. We provide an overview of the most relevant research on intercellular interactions of microglia to evaluate their potential to study brain pathologies.
Collapse
Affiliation(s)
- Angelica Maria Sabogal-Guaqueta
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Teresa Mitchell-Garcia
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Jasmijn Hunneman
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Daniëlle Voshart
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Arun Thiruvalluvan
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Faculty of Science and Engineering, Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences, Section of Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Lara Barazzuol
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, The Netherlands; Department Pathology and Medical biology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
5
|
Bhardwaj JS, Paliwal S, Singhvi G, Taliyan R. Immunological challenges and opportunities in glioblastoma multiforme: A comprehensive view from immune system lens. Life Sci 2024; 357:123089. [PMID: 39362586 DOI: 10.1016/j.lfs.2024.123089] [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: 01/28/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Glioblastoma multiforme (GBM), also known as grade IV astrocytoma, is the most common and deadly brain tumour. It has a poor prognosis and a low survival rate. GBM cells' immunological escape mechanism helps them resist advanced multimodal therapy. In physiological homeostasis, brain astrocytes and microglia suppress infections and clear the potential pathogen from the system. However, in severe pathological conditions like cancer, the immune response fails to eliminate mutated and rapidly over-proliferating GBM cells. The malignant cells' interactions with immune cells and the neoplasm's immunosuppressive environment enable the avoidance and their clearance. Immunotherapy efficiently addresses these difficulties, as shown by sufficient evidence. This review discusses how GBM cells inhibit and elude the immune system. These include MHC molecule expression alteration and PD-L1 and CTLA-4 immune checkpoint overexpression. Without co-stimulation, these changes induce effector T-cell tolerance and anergy. The review also covers how MDSCs, TAMs, Herpes Virus Entry Mediators, and Human cytomegalovirus protein decrease the effector immune response against glioblastoma. The latter part discusses various therapies that are available in the market or under clinical trials which revolves around combating resistance against the available multimodal therapies. The recent trends indicate that there are various monoclonal antibodies and peptide-based vaccines that can be utilized to overcome the immune evasion technique harbored by GBM cells. A strategic development of Immunotherapy considering these hallmarks of immune evasion may help in designing a therapy that may prove to be effective in killing the GBM cells thereby, improving the overall survival of GBM-affected patients.
Collapse
Affiliation(s)
- Jayant Singh Bhardwaj
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Shivangi Paliwal
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| |
Collapse
|
6
|
Wu J, Xu X, Zhang S, Li M, Qiu Y, Lu G, Zheng Z, Huang H. Plastic Events of the Vestibular Nucleus: the Initiation of Central Vestibular Compensation. Mol Neurobiol 2024; 61:9680-9693. [PMID: 38689145 DOI: 10.1007/s12035-024-04208-2] [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: 05/19/2023] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
Vestibular compensation is a physiological response of the vestibular organs within the inner ear. This adaptation manifests during consistent exposure to acceleration or deceleration, with the vestibular organs incrementally adjusting to such changes. The molecular underpinnings of vestibular compensation remain to be fully elucidated, yet emerging studies implicate associations with neuroplasticity and signal transduction pathways. Throughout the compensation process, the vestibular sensory neurons maintain signal transmission to the central equilibrium system, facilitating adaptability through alterations in synaptic transmission and neuronal excitability. Notable molecular candidates implicated in this process include variations in ion channels and neurotransmitter profiles, as well as neuronal and synaptic plasticity, metabolic processes, and electrophysiological modifications. This study consolidates the current understanding of the molecular events in vestibular compensation, augments the existing research landscape, and evaluates contemporary therapeutic strategies. Furthermore, this review posits potential avenues for future research that could enhance our comprehension of vestibular compensation mechanisms.
Collapse
Affiliation(s)
- Junyu Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Xue Xu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Minping Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhihui Zheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Zhang K, Yang Z, Yang Z, Du L, Zhou Y, Fu S, Wang X, Liu D, He X. Targeting microglial GLP1R in epilepsy: A novel approach to modulate neuroinflammation and neuronal apoptosis. Eur J Pharmacol 2024; 981:176903. [PMID: 39154823 DOI: 10.1016/j.ejphar.2024.176903] [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: 05/08/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Epilepsy is a prevalent disorder of the central nervous system. Approximately, one-third of patients show resistance to pharmacological interventions. The pathogenesis of epilepsy is complex, and neuronal apoptosis plays a critical role. Aberrantly reactive astrocytes, induced by cytokine release from activated microglia, may lead to neuronal apoptosis. This study investigated the role of glucagon-like peptide 1 receptor (GLP1R) in microglial activation in epilepsy and its impact on astrocyte-mediated neurotoxicity. METHODS We used human hippocampal tissue from patients with temporal lobe epilepsy and a pilocarpine-induced epileptic mouse model to assess neurobiological changes in epilepsy. BV2 microglial cells and primary astrocytes were used to evaluate cytokine release and astrocyte activation in vitro. The involvement of GLP1R was explored using the GLP1R agonist, Exendin-4 (Ex-4). RESULTS Our findings indicated that reduced GLP1R expression in hippocampal microglia in both epileptic mouse models and human patients, correlated with increased cytokine release and astrocyte activation. Ex-4 treatment restored microglial homeostasis, decreased cytokine secretion, and reduced astrocyte activation, particularly of the A1 phenotype. These changes were associated with a reduction in neuronal apoptosis. In addition, Ex-4 treatment significantly decreased the frequency and duration of seizures in epileptic mice. CONCLUSIONS This study highlights the crucial role of microglial GLP1R in epilepsy pathophysiology. GLP1R downregulation contributes to microglial- and astrocyte-mediated neurotoxicity, exacerbating neuronal death and seizures. Activation of GLP1R with Ex-4 has emerged as a promising therapeutic strategy to reduce neuroinflammation, protect neuronal cells, and control seizures in epilepsy. This study provides a foundation for developing novel antiepileptic therapies targeting microglial GLP1R, with the potential to improve outcomes in patients with epilepsy.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Zhuanyi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Liangchao Du
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Yu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Shiyu Fu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Xiaoyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China
| | - Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
| | - Xinghui He
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
8
|
Wu J, Ren R, Chen T, Su LD, Tang T. Neuroimmune and neuroinflammation response for traumatic brain injury. Brain Res Bull 2024; 217:111066. [PMID: 39241894 DOI: 10.1016/j.brainresbull.2024.111066] [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: 06/15/2024] [Revised: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Traumatic brain injury (TBI) is one of the major diseases leading to mortality and disability, causing a serious disease burden on individuals' ordinary lives as well as socioeconomics. In primary injury, neuroimmune and neuroinflammation are both responsible for the TBI. Besides, extensive and sustained injury induced by neuroimmune and neuroinflammation also prolongs the course and worsens prognosis of TBI. Therefore, this review aims to explore the role of neuroimmune, neuroinflammation and factors associated them in TBI as well as the therapies for TBI. Thus, we conducted by searching PubMed, Scopus, and Web of Science databases for articles published between 2010 and 2023. Keywords included "traumatic brain injury," "neuroimmune response," "neuroinflammation," "astrocytes," "microglia," and "NLRP3." Articles were selected based on relevance and quality of evidence. On this basis, we provide the cellular and molecular mechanisms of TBI-induced both neuroimmune and neuroinflammation response, as well as the different factors affecting them, are introduced based on physiology of TBI, which supply a clear overview in TBI-induced chain-reacting, for a better understanding of TBI and to offer more thoughts on the future therapies for TBI.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| | - Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
9
|
Zhang S, Gao Z, Feng L, Li M. Prevention and Treatment Strategies for Alzheimer's Disease: Focusing on Microglia and Astrocytes in Neuroinflammation. J Inflamm Res 2024; 17:7235-7259. [PMID: 39421566 PMCID: PMC11484773 DOI: 10.2147/jir.s483412] [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: 06/19/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease characterized by its insidious onset and progressive development, making it the most common form of dementia. Despite its prevalence, the exact causes and mechanisms responsible for AD remain unclear. Recent studies have highlighted that inflammation in the central nervous system (CNS) plays a crucial role in both the initiation and progression of AD. Neuroinflammation, an immune response within the CNS triggered by glial cells in response to various stimuli, such as nerve injury, infection, toxins, or autoimmune reactions, has emerged as a significant factor alongside amyloid deposition and neurofibrillary tangles (NFTs) commonly associated with AD. This article aims to provide an overview of the most recent research regarding the involvement of neuroinflammation in AD, with a particular focus on elucidating the specific mechanisms involving microglia and astrocytes. By exploring these intricate processes, a new theoretical framework can be established to further probe the impact of neuroinflammation on the development and progression of AD. Through a deeper understanding of these underlying mechanisms, potential targets for therapeutic interventions and novel treatment strategies can be identified in the ongoing battle against AD.
Collapse
Affiliation(s)
- Shenghao Zhang
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Zhejianyi Gao
- Department of Orthopaedics, Fushun Hospital of Chinese Medicine, Fushun, Liaoning Province, 113008, People’s Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
- Shandong Key Laboratory of TCM Multi-Targets Intervention and Disease Control, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, 271000, People’s Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| |
Collapse
|
10
|
Mo X, Cheng R, Shen L, Liu N, Sun Y, Lin S, Jiang G, Li X, Peng X, Zhang Y, Liao Y, Yan H, Liu L. Yeast β-glucan alleviates high-fat diet-induced Alzheimer's disease-like pathologies in rats via the gut-brain axis. Int J Biol Macromol 2024; 278:134939. [PMID: 39179066 DOI: 10.1016/j.ijbiomac.2024.134939] [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: 03/13/2024] [Revised: 05/15/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Targeting the gut microbiota may be an emerging strategy for the prevention and treatment of Alzheimer's disease (AD). Macro-molecular yeast β-glucan (BG), derived from the yeast of Saccharomyces cerevisiae, regulates the gut microbiota. This study aimed to investigate the effect and mechanism of long-term BG in high-fat diet (HFD)-induced AD-like pathologies from the perspective of the gut microbiota. Here, we found that 80 weeks of BG treatment ameliorated HFD-induced cognitive dysfunction in rats. In the hippocampus, BG alleviated HFD-induced the activation of astrocytes, microglia, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome pathway, and AD-like pathologies. BG modulated gut dysbiosis through increasing the levels of beneficial bacteria and short-chain fatty acids (SCFAs). BG also attenuated HFD-induced gut barrier impairment. Correlation analysis revealed a close relationship among microbiota, SCFAs, and AD-like pathologies. Furthermore, the fecal microbiota of BG-treated rats and SCFAs treatment mitigated AD-like pathologies via the NLRP3 inflammasome pathway in HFD-fed aged rats. These results suggested that long-term BG promotes the production of SCFAs derived from gut microbiota, which further inhibits NLRP3 inflammasome-mediated neuroinflammation, thereby alleviating HFD-induced AD-like pathologies in rats. BG may become a new strategy for targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoxing Mo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Ruijie Cheng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Lihui Shen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Nian Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Yunhong Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Shan Lin
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Guanhua Jiang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Xiaoqin Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Xiaobo Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Yan Zhang
- The Hubei Provincial Key Laboratory of Yeast Function, Yichang 443003, China.
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| |
Collapse
|
11
|
Lopez-Ortiz AO, Eyo UB. Astrocytes and microglia in the coordination of CNS development and homeostasis. J Neurochem 2024; 168:3599-3614. [PMID: 37985374 PMCID: PMC11102936 DOI: 10.1111/jnc.16006] [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: 08/31/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Glia have emerged as important architects of central nervous system (CNS) development and maintenance. While traditionally glial contributions to CNS development and maintenance have been studied independently, there is growing evidence that either suggests or documents that glia may act in coordinated manners to effect developmental patterning and homeostatic functions in the CNS. In this review, we focus on astrocytes, the most abundant glia in the CNS, and microglia, the earliest glia to colonize the CNS highlighting research that documents either suggestive or established coordinated actions by these glial cells in various CNS processes including cell and/or debris clearance, neuronal survival and morphogenesis, synaptic maturation, and circuit function, angio-/vasculogenesis, myelination, and neurotransmission. Some molecular mechanisms underlying these processes that have been identified are also described. Throughout, we categorize the available evidence as either suggestive or established interactions between microglia and astrocytes in the regulation of the respective process and raise possible avenues for further research. We conclude indicating that a better understanding of coordinated astrocyte-microglial interactions in the developing and mature brain holds promise for developing effective therapies for brain pathologies where these processes are perturbed.
Collapse
Affiliation(s)
- Aída Oryza Lopez-Ortiz
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Ukpong B Eyo
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
12
|
Zhao Y, Huang Y, Cao Y, Yang J. Astrocyte-Mediated Neuroinflammation in Neurological Conditions. Biomolecules 2024; 14:1204. [PMID: 39456137 PMCID: PMC11505625 DOI: 10.3390/biom14101204] [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: 08/09/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Astrocytes are one of the key glial types of the central nervous system (CNS), accounting for over 20% of total glial cells in the brain. Extensive evidence has established their indispensable functions in the maintenance of CNS homeostasis, as well as their broad involvement in neurological conditions. In particular, astrocytes can participate in various neuroinflammatory processes, e.g., releasing a repertoire of cytokines and chemokines or specific neurotrophic factors, which result in both beneficial and detrimental effects. It has become increasingly clear that such astrocyte-mediated neuroinflammation, together with its complex crosstalk with other glial cells or immune cells, designates neuronal survival and the functional integrity of neurocircuits, thus critically contributing to disease onset and progression. In this review, we focus on the current knowledge of the neuroinflammatory responses of astrocytes, summarizing their common features in neurological conditions. Moreover, we highlight several vital questions for future research that promise novel insights into diagnostic or therapeutic strategies against those debilitating CNS diseases.
Collapse
Affiliation(s)
- Yanxiang Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- The Affiliated High School, Peking University, Beijing 100080, China
| | - Yingying Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ying Cao
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Third Hospital Cancer Center, Beijing 100191, China
| |
Collapse
|
13
|
Liu SL, Tang Y. Astrocytic adenosine A 1 receptors: a new potential target for treating sepsis-associated encephalopathy. Purinergic Signal 2024:10.1007/s11302-024-10049-4. [PMID: 39240445 DOI: 10.1007/s11302-024-10049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Affiliation(s)
- Si-Le Liu
- International Joint Research Centre On Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Yong Tang
- International Joint Research Centre On Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| |
Collapse
|
14
|
Zhang J, Hu D, Li L, Qu D, Shi W, Xie L, Jiang Q, Li H, Yu T, Qi C, Fu H. M2 Microglia-derived Exosomes Promote Spinal Cord Injury Recovery in Mice by Alleviating A1 Astrocyte Activation. Mol Neurobiol 2024; 61:7009-7025. [PMID: 38367135 DOI: 10.1007/s12035-024-04026-6] [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: 10/23/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024]
Abstract
M2 microglia transplantation has previously demonstrated beneficial effects on spinal cord injury (SCI) by regulating neuroinflammation and enhancing neuronal survival. Exosomes (EXOs), secreted by almost all cell types, embody partial functions and properties of their parent cells. However, the effect of M2 microglia-derived EXOs (M2-EXOs) on SCI recovery and the underlying molecular mechanisms remain unclear. In this study, we isolated M2-EXOs and intravenously introduced them into mice with SCI. Considering the reciprocal communication between microglia and astroglia in both healthy and injured central nervous systems (CNSs), we subsequently focused on the influence of M2-EXOs on astrocyte phenotype regulation. Our findings indicated that M2-EXOs promoted neuron survival and axon preservation, reduced the lesion area, inhibited A1 astrocyte activation, and improved motor function recovery in SCI mice. Moreover, they inhibited the nuclear translocation of p65 and the activation of the NF-κB signalling pathway in A1 astrocytes. Therefore, our research suggests that M2-EXOs mitigate the activation of neurotoxic A1 astrocytes by inhibiting the NF-κB signalling pathway, thereby improving spinal tissue preservation and motor function recovery following SCI. This positions M2-EXOs as a promising therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Die Hu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Liping Li
- Department of Bone Surgery, Qingdao Central Hospital, Qingdao, 266000, China
| | - Di Qu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Weipeng Shi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Lei Xie
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Qi Jiang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Medical Department of, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Haifeng Li
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Tengbo Yu
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
- Institute of Sports Medicine and Health, Qingdao University, Qingdao, 266000, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
15
|
Pascoal T, Rohden F, Ferreira P, Bellaver B, Ferrari-Souza JP, Aguzzoli C, Soares C, Abbas S, Zalzale H, Povala G, Lussier F, Leffa D, Bauer-Negrini G, Rahmouni N, Tissot C, Therriault JT, Servaes S, Stevenson J, Benedet A, Ashton N, Karikari T, Tudorascu D, Zetterberg H, Blennow K, Zimmer E, Souza D, Rosa-Neto P. Glial reactivity is linked to synaptic dysfunction across the aging and Alzheimer's disease spectrum. RESEARCH SQUARE 2024:rs.3.rs-4782732. [PMID: 39184102 PMCID: PMC11343173 DOI: 10.21203/rs.3.rs-4782732/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Previous studies have shown that glial and neuronal changes may trigger synaptic dysfunction in Alzheimer's disease(AD). However, the link between glial and neuronal markers and synaptic abnormalities in the living brain is poorly understood. Here, we investigated the association between biomarkers of astrocyte and microglial reactivity and synaptic dysfunction in 478 individuals across the aging and AD spectrum from two cohorts with available CSF measures of amyloid-β(Aβ), phosphorylated tau(pTau181), astrocyte reactivity(GFAP), microglial activation(sTREM2), and synaptic biomarkers(GAP43 and neurogranin). Elevated CSF GFAP levels were linked to presynaptic and postsynaptic dysfunction, regardless of cognitive status or Aβ presence. CSF sTREM2 levels were associated with presynaptic biomarkers in cognitively unimpaired and impaired Aβ + individuals and postsynaptic biomarkers in cognitively impaired Aβ + individuals. Notably, CSF pTau181 levels mediated all associations between GFAP or sTREM2 levels and synaptic dysfunction biomarkers. These results suggest that neuronal-related synaptic biomarkers could be used in clinical trials targeting glial reactivity in AD.
Collapse
|
16
|
Brockie S, Zhou C, Fehlings MG. Resident immune responses to spinal cord injury: role of astrocytes and microglia. Neural Regen Res 2024; 19:1678-1685. [PMID: 38103231 PMCID: PMC10960308 DOI: 10.4103/1673-5374.389630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury can be traumatic or non-traumatic in origin, with the latter rising in incidence and prevalence with the aging demographics of our society. Moreover, as the global population ages, individuals with co-existent degenerative spinal pathology comprise a growing number of traumatic spinal cord injury cases, especially involving the cervical spinal cord. This makes recovery and treatment approaches particularly challenging as age and comorbidities may limit regenerative capacity. For these reasons, it is critical to better understand the complex milieu of spinal cord injury lesion pathobiology and the ensuing inflammatory response. This review discusses microglia-specific purinergic and cytokine signaling pathways, as well as microglial modulation of synaptic stability and plasticity after injury. Further, we evaluate the role of astrocytes in neurotransmission and calcium signaling, as well as their border-forming response to neural lesions. Both the inflammatory and reparative roles of these cells have eluded our complete understanding and remain key therapeutic targets due to their extensive structural and functional roles in the nervous system. Recent advances have shed light on the roles of glia in neurotransmission and reparative injury responses that will change how interventions are directed. Understanding key processes and existing knowledge gaps will allow future research to effectively target these cells and harness their regenerative potential.
Collapse
Affiliation(s)
- Sydney Brockie
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Cindy Zhou
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Wang Y, Li H, Sun H, Xu C, Sun H, Wei W, Song J, Jia F, Zhong D, Li G. A2 reactive astrocyte-derived exosomes alleviate cerebral ischemia-reperfusion injury by delivering miR-628. J Cell Mol Med 2024; 28:e70004. [PMID: 39159174 PMCID: PMC11332600 DOI: 10.1111/jcmm.70004] [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: 10/10/2023] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
Ischemia and hypoxia activate astrocytes into reactive types A1 and A2, which play roles in damage and protection, respectively. However, the function and mechanism of A1 and A2 astrocyte exosomes are unknown. After astrocyte exosomes were injected into the lateral ventricle, infarct volume, damage to the blood-brain barrier (BBB), apoptosis and the expression of microglia-related proteins were measured. The dual luciferase reporter assay was used to detect the target genes of miR-628, and overexpressing A2-Exos overexpressed and knocked down miR-628 were constructed. qRT-PCR, western blotting and immunofluorescence staining were subsequently performed. A2-Exos obviously reduced the infarct volume, damage to the BBB and apoptosis and promoted M2 microglial polarization. RT-PCR showed that miR-628 was highly expressed in A2-Exos. Dual luciferase reporter assays revealed that NLRP3, S1PR3 and IRF5 are target genes of miR-628. After miR-628 was overexpressed or knocked down, the protective effects of A2-Exos increased or decreased, respectively. A2-Exos reduced pyroptosis and BBB damage and promoted M2 microglial polarization through the inhibition of NLRP3, S1PR3 and IRF5 via the delivery of miR-628. This study explored the mechanism of action of A2-Exos and provided new therapeutic targets and concepts for treating cerebral ischemia.
Collapse
Affiliation(s)
- Yingju Wang
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - He Li
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Hanwen Sun
- Department of EmergencyRui Jin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Chen Xu
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Hongxue Sun
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Wan Wei
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Jihe Song
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Feihong Jia
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Di Zhong
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Guozhong Li
- Department of NeurologyThe First Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
- Department of NeurologyHeilongjiang Provincial HospitalHarbinHeilongjiangPeople's Republic of China
| |
Collapse
|
18
|
Lee H, Pearse RV, Lish AM, Pan C, Augur ZM, Terzioglu G, Gaur P, Liao M, Fujita M, Tio ES, Duong DM, Felsky D, Seyfried NT, Menon V, Bennett DA, De Jager PL, Young-Pearse TL. Contributions of genetic variation in astrocytes to cell and molecular mechanisms of risk and resilience to late onset Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605928. [PMID: 39211227 PMCID: PMC11361137 DOI: 10.1101/2024.07.31.605928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Reactive astrocytes are associated with Alzheimer's disease (AD), and several AD genetic risk variants are associated with genes highly expressed in astrocytes. However, the contribution of genetic risk within astrocytes to cellular processes relevant to the pathogenesis of AD remains ill-defined. Here we present a resource for studying AD genetic risk in astrocytes using a large collection of induced pluripotent stem cell (iPSC) lines from deeply phenotyped individuals with a range of neuropathological and cognitive outcomes. IPSC lines from forty-four individuals were differentiated into astrocytes followed by unbiased molecular profiling using RNA sequencing and tandem mass tag-mass spectrometry. We demonstrate the utility of this resource in examining gene- and pathway-level associations with clinical and neuropathological traits, as well as in analyzing genetic risk and resilience factors through parallel analyses of iPSC-astrocytes and brain tissue from the same individuals. Our analyses reveal that genes and pathways altered in iPSC-derived astrocytes from AD individuals are concordantly dysregulated in AD brain tissue. This includes increased prefoldin proteins, extracellular matrix factors, COPI-mediated trafficking components and reduced proteins involved in cellular respiration and fatty acid oxidation. Additionally, iPSC-derived astrocytes from individuals resilient to high AD neuropathology show elevated basal levels of interferon response proteins and increased secretion of interferon gamma. Correspondingly, higher polygenic risk scores for AD are associated with lower levels of interferon response proteins. This study establishes an experimental system that integrates genetic information with a heterogeneous set of iPSCs to identify genetic contributions to molecular pathways affecting AD risk and resilience.
Collapse
|
19
|
Lu CL, Ren J, Cao X. An Astroglial Basis of Major Depressive Disorder: Molecular, Cellular, and Circuit Features. Biol Psychiatry 2024:S0006-3223(24)01484-7. [PMID: 39084500 DOI: 10.1016/j.biopsych.2024.07.017] [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: 01/28/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
Major depressive disorder is a common psychiatric disorder and a leading cause of disability worldwide. Astrocytes play a role in the maintenance of the function of the central nervous system, both physiologically and pathologically. Accumulated evidence indicates that the astrocyte is an important contributor to the pathophysiology of major depressive disorder including blood-brain barrier integrity, gap junctions, gliotransmission, glutamate homeostasis, and energy metabolism. Here, we comprehensively summarize an astroglial basis for major depressive disorder based on molecular, cellular, and circuit properties, suggesting that astrocytes appear to be highly sensitive to stress and are likely to be uniquely positioned to integrate peripheral and central stress responses.
Collapse
Affiliation(s)
- Cheng-Lin Lu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China; Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jing Ren
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiong Cao
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People's Republic of China; Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Guo Q, Gobbo D, Zhao N, Zhang H, Awuku NO, Liu Q, Fang LP, Gampfer TM, Meyer MR, Zhao R, Bai X, Bian S, Scheller A, Kirchhoff F, Huang W. Adenosine triggers early astrocyte reactivity that provokes microglial responses and drives the pathogenesis of sepsis-associated encephalopathy in mice. Nat Commun 2024; 15:6340. [PMID: 39068155 PMCID: PMC11283516 DOI: 10.1038/s41467-024-50466-y] [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: 11/10/2023] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Molecular pathways mediating systemic inflammation entering the brain parenchyma to induce sepsis-associated encephalopathy (SAE) remain elusive. Here, we report that in mice during the first 6 hours of peripheral lipopolysaccharide (LPS)-evoked systemic inflammation (6 hpi), the plasma level of adenosine quickly increased and enhanced the tone of central extracellular adenosine which then provoked neuroinflammation by triggering early astrocyte reactivity. Specific ablation of astrocytic Gi protein-coupled A1 adenosine receptors (A1ARs) prevented this early reactivity and reduced the levels of inflammatory factors (e.g., CCL2, CCL5, and CXCL1) in astrocytes, thereby alleviating microglial reaction, ameliorating blood-brain barrier disruption, peripheral immune cell infiltration, neuronal dysfunction, and depression-like behaviour in the mice. Chemogenetic stimulation of Gi signaling in A1AR-deficent astrocytes at 2 and 4 hpi of LPS injection could restore neuroinflammation and depression-like behaviour, highlighting astrocytes rather than microglia as early drivers of neuroinflammation. Our results identify early astrocyte reactivity towards peripheral and central levels of adenosine as an important pathway driving SAE and highlight the potential of targeting A1ARs for therapeutic intervention.
Collapse
Affiliation(s)
- Qilin Guo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Na Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology, University of Saarland, 66421, Homburg, Germany
| | - Hong Zhang
- Biophysics, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Nana-Oye Awuku
- Molecular Neurophysiology, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Qing Liu
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
| | - Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Tanja M Gampfer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), University of Saarland, 66421, Homburg, Germany
| | - Markus R Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), University of Saarland, 66421, Homburg, Germany
| | - Renping Zhao
- Biophysics, CIPMM, University of Saarland, 66421, Homburg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Shan Bian
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421, Homburg, Germany.
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, 66421, Homburg, Germany.
| |
Collapse
|
21
|
Maccioni L, Michelle CM, Brusaferri L, Silvestri E, Bertoldo A, Schubert JJ, Nettis MA, Mondelli V, Howes O, Turkheimer FE, Bottlaender M, Bodini B, Stankoff B, Loggia ML, Veronese M. A blood-free modeling approach for the quantification of the blood-to-brain tracer exchange in TSPO PET imaging. Front Neurosci 2024; 18:1395769. [PMID: 39104610 PMCID: PMC11299498 DOI: 10.3389/fnins.2024.1395769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Recent evidence suggests the blood-to-brain influx rate (K1 ) in TSPO PET imaging as a promising biomarker of blood-brain barrier (BBB) permeability alterations commonly associated with peripheral inflammation and heightened immune activity in the brain. However, standard compartmental modeling quantification is limited by the requirement of invasive and laborious procedures for extracting an arterial blood input function. In this study, we validate a simplified blood-free methodologic framework for K1 estimation by fitting the early phase tracer dynamics using a single irreversible compartment model and an image-derived input function (1T1K-IDIF). Methods The method is tested on a multi-site dataset containing 177 PET studies from two TSPO tracers ([11C]PBR28 and [18F]DPA714). Firstly, 1T1K-IDIF K1 estimates were compared in terms of both bias and correlation with standard kinetic methodology. Then, the method was tested on an independent sample of [11C]PBR28 scans before and after inflammatory interferon-α challenge, and on test-retest dataset of [18F]DPA714 scans. Results Comparison with standard kinetic methodology showed good-to-excellent intra-subject correlation for regional 1T1K-IDIF-K1 (ρintra = 0.93 ± 0.08), although the bias was variable depending on IDIF ability to approximate blood input functions (0.03-0.39 mL/cm3/min). 1T1K-IDIF-K1 unveiled a significant reduction of BBB permeability after inflammatory interferon-α challenge, replicating results from standard quantification. High intra-subject correlation (ρ = 0.97 ± 0.01) was reported between K1 estimates of test and retest scans. Discussion This evidence supports 1T1K-IDIF as blood-free alternative to assess TSPO tracers' unidirectional blood brain clearance. K1 investigation could complement more traditional measures in TSPO studies, and even allow further mechanistic insight in the interpretation of TSPO signal.
Collapse
Affiliation(s)
- Lucia Maccioni
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Carranza Mellana Michelle
- Department of Information Engineering, University of Padova, Padova, Italy
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Ludovica Brusaferri
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
- Computer Science and Informatics, School of Engineering, London South Bank University, London, United Kingdom
| | - Erica Silvestri
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alessandra Bertoldo
- Department of Information Engineering, University of Padova, Padova, Italy
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Julia J. Schubert
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Maria A. Nettis
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valeria Mondelli
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Oliver Howes
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Federico E. Turkheimer
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Michel Bottlaender
- BioMaps, Service Hospitalier Frédéric Joliot CEA, CNRS Inserm, Université Paris-Saclay, Orsay, France
| | - Benedetta Bodini
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Bruno Stankoff
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, Paris, France
| | - Marco L. Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Mattia Veronese
- Department of Information Engineering, University of Padova, Padova, Italy
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| |
Collapse
|
22
|
Tao B, Gong W, Xu C, Ma Z, Mei J, Chen M. The relationship between hypoxia and Alzheimer's disease: an updated review. Front Aging Neurosci 2024; 16:1402774. [PMID: 39086755 PMCID: PMC11288848 DOI: 10.3389/fnagi.2024.1402774] [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: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, and the most prevalent form of dementia. The main hallmarks for the diagnosis of AD are extracellular amyloid-beta (Aβ) plaque deposition and intracellular accumulation of highly hyperphosphorylated Tau protein as neurofibrillary tangles. The brain consumes more oxygen than any other organs, so it is more easily to be affected by hypoxia. Hypoxia has long been recognized as one of the possible causes of AD and other neurodegenerative diseases, but the exact mechanism has not been clarified. In this review, we will elucidate the connection between hypoxia-inducible factors-1α and AD, including its contribution to AD and its possible protective effects. Additionally, we will discuss the relationship between oxidative stress and AD as evidence show that oxidative stress acts on AD-related pathogenic factors such as mitochondrial dysfunction, Aβ deposition, inflammation, etc. Currently, there is no cure for AD. Given the close association between hypoxia, oxidative stress, and AD, along with current research on the protective effects of antioxidants against AD, we speculate that antioxidants could be a potential therapeutic approach for AD and worth further study.
Collapse
Affiliation(s)
- Borui Tao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- The First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Wei Gong
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengyuan Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhihui Ma
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinyu Mei
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
23
|
Jin M, Ma Z, Zhang H, Papetti AV, Dang R, Stillitano AC, Goldman SA, Jiang P. Co-Transplantation-Based Human-Mouse Chimeric Brain Models to Study Human Glial-Glial and Glial-Neuronal Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601990. [PMID: 39005270 PMCID: PMC11244967 DOI: 10.1101/2024.07.03.601990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Human-mouse chimeric brain models, generated by transplanting human induced pluripotent stem cell (hiPSC)-derived neural cells, are valuable for studying the development and function of human neural cells in vivo. Understanding glial-glial and glial-neuronal interactions is essential for unraveling the complexities of brain function and developing treatments for neurological disorders. To explore these interactions between human neural cells within an intact brain environment, we employe a co-transplantation strategy involving the engraftment of hiPSC-derived neural progenitor cells along with primitive macrophage progenitors into the neonatal mouse brain. This approach creates human-mouse chimeric brains containing human microglia, macroglia (astroglia and oligodendroglia), and neurons. Using super-resolution imaging and 3D reconstruction techniques, we examine the dynamics between human neurons and glia, unveiling human microglia engulfing immature human neurons, microglia pruning synapses of human neurons, and significant interactions between human oligodendrocytes and neurons. Single-cell RNA sequencing analysis of the chimeric brain uncovers a close recapitulation of the human glial progenitor cell population, along with a dynamic stage in astroglial development that mirrors the processes found in the human brain. Furthermore, cell-cell communication analysis highlights significant neuronal-glial and glial-glial interactions, especially the interaction between adhesion molecules neurexins and neuroligins. This innovative co-transplantation model opens up new avenues for exploring the complex pathophysiological mechanisms underlying human neurological diseases. It holds particular promise for studying disorders where glial-neuronal interactions and non-cell-autonomous effects play crucial roles.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Haiwei Zhang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Ava V. Papetti
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Rui Dang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Lead Contact
| |
Collapse
|
24
|
Liddelow SA, Olsen ML, Sofroniew MV. Reactive Astrocytes and Emerging Roles in Central Nervous System (CNS) Disorders. Cold Spring Harb Perspect Biol 2024; 16:a041356. [PMID: 38316554 PMCID: PMC11216178 DOI: 10.1101/cshperspect.a041356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute, NYU School of Medicine, New York, New York 10016, USA
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, New York 10016, USA
- Department of Ophthalmology, NYU School of Medicine, New York, New York 10016, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
25
|
Chen K, Wei X, Zhang W, Wang R, Wang Y, Yang L. Bone morphogenetic protein 4 derived from the cerebrospinal fluid in patients with postherpetic neuralgia induces allodynia via the crosstalk between microglia and astrocyte. Brain Behav Immun 2024; 119:836-850. [PMID: 38735405 DOI: 10.1016/j.bbi.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION During postherpetic neuralgia (PHN), the cerebral spinal fluid (CSF) possesses the capability to trigger glial activation and inflammation, yet the specific changes in its composition remain unclear. Recent findings from our research indicate elevations of central bone morphogenetic protein 4 (BMP4) during neuropathic pain (NP), serving as an independent modulator of glial cells. Herein, the aim of the present study is to test the CSF-BMP4 expressions and its role in the glial modulation in the process of PHN. METHODS CSF samples were collected from both PHN patients and non-painful individuals (Control) to assess BMP4 and its antagonist Noggin levels. Besides, intrathecal administration of both CSF types was conducted in normal rats to evaluate the impact on pain behavior, glial activity, and inflammation.; Additionally, both Noggin and STAT3 antagonist-Stattic were employed to treat the PHN-CSF or exogenous BMP4 challenged cultured astrocytes to explore downstream signals. Finally, microglial depletion was performed prior to the PHN-CSF intervention so as to elucidate the microglia-astrocyte crosstalk. RESULTS BMP4 levels were significantly higher in PHN-CSF compared to Control-CSF (P < 0.001), with a positive correlation with pain duration (P < 0.05, r = 0.502). Comparing with the Control-CSF producing moderate paw withdrawal threshold (PWT) decline and microglial activation, PHN-CSF further exacerbated allodynia and triggered both microglial and astrocytic activation (P < 0.05). Moreover, PHN-CSF rather than Control-CSF evoked microglial proliferation and pro-inflammatory transformation, reinforced iron storage, and activated astrocytes possibly through both SMAD159 and STAT3 signaling, which were all mitigated by the Noggin application (P < 0.05). Next, both Noggin and Stattic effectively attenuated BMP4-induced GFAP and IL-6 upregulation, as well as SMAD159 and STAT3 phosphorylation in the cultured astrocytes (P < 0.05). Finally, microglial depletion diminished PHN-CSF induced astrogliosis, inflammation and endogenous BMP4 expression (P < 0.05). CONCLUSION Our study highlights the role of CSF-BMP4 elevation in glial activation and allodynia during PHN, suggesting a potential therapeutic avenue for future exploration.
Collapse
Affiliation(s)
- Kai Chen
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China
| | - Xiaojin Wei
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China
| | - Wenjuan Zhang
- Department of the Laboratory, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruixuan Wang
- Bourns Engineering, The University of California, Riverside, CA 92521, USA
| | - Yaping Wang
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China.
| | - Lin Yang
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China.
| |
Collapse
|
26
|
Ohbuchi M, Shibuta M, Tetsuka K, Sasaki-Iwaoka H, Oishi M, Shimizu F, Nagasaka Y. Modeling of Blood-Brain Barrier (BBB) Dysfunction and Immune Cell Migration Using Human BBB-on-a-Chip for Drug Discovery Research. Int J Mol Sci 2024; 25:6496. [PMID: 38928202 PMCID: PMC11204321 DOI: 10.3390/ijms25126496] [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: 03/31/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Blood-brain barrier (BBB) dysfunction is a key feature in neuroimmunological and neurodegenerative diseases. In this study, we developed a microfluidic human BBB-on-a-chip to model barrier dysfunction and immune cell migration using immortalized TY10 brain endothelial cells, pericytes, and astrocytes. It was found that immortalized TY10 brain endothelial cells developed a microvascular structure under flow. Pericytes were localized on the basal side surrounding the TY10 microvascular structure, showing an in vivo-like structure. Barrier integrity increased under co-culture with pericytes. In addition, both ethylenediaminetetraacetic acid (EDTA) and anti-Claudin-5 (CLDN5) neutralizing antibody caused a decrease in the transendothelial electrical resistance (TEER). EDTA caused the leakage of 20 kDa dextran, suggesting different effects on the BBB based on the mechanism of action, whereas anti-CLDN5 antibody did not cause leakage. In the tri-culture model, human T cells migrated through endothelial vessels towards basal C-X-C motif chemokine ligand 12 (CXCL12). The live-imaging analysis confirmed the extravasation of fluorescence-labelled T cells in a CXCL12-concentration- and time-dependent manner. Our BBB model had an in vivo-like structure and successfully represented barrier dysfunction and transendothelial T cell migration. In addition, our study suggests that the inhibition of CLDN5 attenuates the BBB in humans. This platform has various potential uses in relation to the BBB in both drug discovery research and in elucidating the mechanisms of central nervous system diseases.
Collapse
Affiliation(s)
- Masato Ohbuchi
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| | - Mayu Shibuta
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| | - Kazuhiro Tetsuka
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| | - Haruna Sasaki-Iwaoka
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| | - Masayo Oishi
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube 755-8505, Yamaguchi, Japan;
| | - Yasuhisa Nagasaka
- Applied Research & Operations, Astellas Pharma Inc., Tsukuba 305-8585, Ibaraki, Japan; (M.S.); (K.T.); (H.S.-I.); (M.O.); (Y.N.)
| |
Collapse
|
27
|
Lee KS, Yoon SH, Hwang I, Ma JH, Yang E, Kim RH, Kim E, Yu JW. Hyperglycemia enhances brain susceptibility to lipopolysaccharide-induced neuroinflammation via astrocyte reprogramming. J Neuroinflammation 2024; 21:137. [PMID: 38802820 PMCID: PMC11131277 DOI: 10.1186/s12974-024-03136-1] [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: 12/17/2023] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Hyperglycemia has been shown to modulate the immune response of peripheral immune cells and organs, but the impact of hyperglycemia on neuroinflammation within the brain remains elusive. In the present study, we provide evidences that streptozotocin (STZ)-induced hyperglycemic condition in mice drives a phenotypic switch of brain astrocytes to a proinflammatory state, and increases brain vulnerability to mild peripheral inflammation. In particular, we found that hyperglycemia led to a significant increase in the astrocyte proliferation as determined by flow cytometric and immunohistochemical analyses of mouse brain. The increased astrocyte proliferation by hyperglycemia was reduced by Glut1 inhibitor BAY-876. Transcriptomic analysis of isolated astrocytes from Aldh1l1CreERT2;tdTomato mice revealed that peripheral STZ injection induced astrocyte reprogramming into proliferative, and proinflammatory phenotype. Additionally, STZ-induced hyperglycemic condition significantly enhanced the infiltration of circulating myeloid cells into the brain and the disruption of blood-brain barrier in response to mild lipopolysaccharide (LPS) administration. Systemic hyperglycemia did not alter the intensity and sensitivity of peripheral inflammation in mice to LPS challenge, but increased the inflammatory potential of brain microglia. In line with findings from mouse experiments, a high-glucose environment intensified the LPS-triggered production of proinflammatory molecules in primary astrocyte cultures. Furthermore, hyperglycemic mice exhibited a significant impairment in cognitive function after mild LPS administration compared to normoglycemic mice as determined by novel object recognition and Y-maze tasks. Taken together, these results demonstrate that hyperglycemia directly induces astrocyte reprogramming towards a proliferative and proinflammatory phenotype, which potentiates mild LPS-triggered inflammation within brain parenchymal regions.
Collapse
Affiliation(s)
- Kyung-Seo Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Hyun Yoon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Hwa Ma
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Euimo Yang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Rebekah Hyeyoon Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eosu Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
28
|
Champagne-Jorgensen K, Gommerman J. Astrocytes ACLYmate to chronic neuroinflammation. Trends Immunol 2024; 45:320-321. [PMID: 38632002 DOI: 10.1016/j.it.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Astrocytes are essential cells of the mammalian central nervous system (CNS), with key roles in development, homeostasis, and disease. Lee and colleagues recently showed that astrocytes can develop epigenetic memory, which enhances proinflammatory responses to subsequent stimulation, potentially driving sustained neurological disease pathology, such as in multiple sclerosis (MS).
Collapse
Affiliation(s)
- Kevin Champagne-Jorgensen
- University of Toronto, Department of Immunology, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Jennifer Gommerman
- University of Toronto, Department of Immunology, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
29
|
Siva Venkatesh IP, Majumdar A, Basu A. Prophylactic Administration of Gut Microbiome Metabolites Abrogated Microglial Activation and Subsequent Neuroinflammation in an Experimental Model of Japanese Encephalitis. ACS Chem Neurosci 2024; 15:1712-1727. [PMID: 38581382 DOI: 10.1021/acschemneuro.4c00028] [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] [Indexed: 04/08/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are gut microbial metabolic derivatives produced during the fermentation of ingested complex carbohydrates. SCFAs have been widely regarded to have a potent anti-inflammatory and neuro-protective role and have implications in several disease conditions, such as, inflammatory bowel disease, type-2 diabetes, and neurodegenerative disorders. Japanese encephalitis virus (JEV), a neurotropic flavivirus, is associated with life threatening neuro-inflammation and neurological sequelae in infected hosts. In this study, we hypothesize that SCFAs have potential in mitigating JEV pathogenesis. Postnatal day 10 BALB/c mice were intraperitoneally injected with either a SCFA mixture (acetate, propionate, and butyrate) or PBS for a period of 7 days, followed by JEV infection. All mice were observed for onset and progression of symptoms. The brain tissue was collected upon reaching terminal illness for further analysis. SCFA-supplemented JEV-infected mice (SCFA + JEV) showed a delayed onset of symptoms, lower hindlimb clasping score, and decreased weight loss and increased survival by 3 days (p < 0.0001) upon infection as opposed to the PBS-treated JEV-infected animals (JEV). Significant downregulation of inflammatory cytokines TNF-α, MCP-1, IL-6, and IFN-Υ in the SCFA + JEV group relative to the JEV-infected control group was observed. Inflammatory mediators, phospho-NF-kB (P-NF-kB) and iba1, showed 2.08 ± 0.1 and 3.132 ± 0.43-fold upregulation in JEV versus 1.19 ± 0.11 and 1.31 ± 0.11-fold in the SCFA + JEV group, respectively. Tissue section analysis exhibited reduced glial activation (JEV group─42 ± 2.15 microglia/ROI; SCFA + JEV group─27.07 ± 1.8 microglia/ROI) in animals that received SCFA supplementation prior to infection as seen from the astrocytic and microglial morphometric analysis. Caspase-3 immunoblotting showed 4.08 ± 1.3-fold upregulation in JEV as compared to 1.03 ± 0.14-fold in the SCFA + JEV group and TUNEL assay showed a reduced cellular death post-JEV infection (JEV-6.4 ± 1.5 cells/ROI and SCFA + JEV-3.7 ± 0.73 cells/ROI). Our study critically contributes to the increasing evidence in support of SCFAs as an anti-inflammatory and neuro-protective agent, we further expand its scope as a potential supplementary intervention in JEV-mediated neuroinflammation.
Collapse
MESH Headings
- Gastrointestinal Microbiome/physiology
- Neuroinflammatory Diseases/drug therapy
- Neuroinflammatory Diseases/immunology
- Neuroinflammatory Diseases/metabolism
- Neuroinflammatory Diseases/microbiology
- Microglia/drug effects
- Microglia/immunology
- Encephalitis, Japanese/drug therapy
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/microbiology
- Encephalitis, Japanese/prevention & control
- Encephalitis, Japanese/virology
- Fatty Acids, Volatile/pharmacology
- Fatty Acids, Volatile/therapeutic use
- Encephalitis Viruses, Japanese/drug effects
- Encephalitis Viruses, Japanese/immunology
- Encephalitis Viruses, Japanese/pathogenicity
- Survival Analysis
- Chemokines/immunology
- Chemokines/metabolism
- Inflammation Mediators/immunology
- Inflammation Mediators/metabolism
- Cytokine Release Syndrome/immunology
- Cytokine Release Syndrome/metabolism
- Cytokine Release Syndrome/prevention & control
- Humans
- Female
- Animals
- Mice
- Apoptosis/drug effects
- Brain/drug effects
- Brain/metabolism
- Brain/virology
- Viral Load/drug effects
- Time Factors
Collapse
Affiliation(s)
| | - Atreye Majumdar
- National Brain Research Centre, Manesar, Haryana 122052, India
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana 122052, India
| |
Collapse
|
30
|
Müller L, Di Benedetto S. Aging brain: exploring the interplay between bone marrow aging, immunosenescence, and neuroinflammation. Front Immunol 2024; 15:1393324. [PMID: 38638424 PMCID: PMC11024322 DOI: 10.3389/fimmu.2024.1393324] [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: 02/28/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Abstract
Aging is a complex process characterized by a myriad of physiological changes, including alterations in the immune system termed immunosenescence. It exerts profound effects on both the bone marrow and the central nervous system, with significant implications for immunosenescence in neurological contexts. Our mini-review explores the complex relationship between bone marrow aging and its impact on immunosenescence, specifically within the context of neurological diseases. The bone marrow serves as a crucial hub for hematopoiesis and immune cell production, yet with age, it undergoes significant alterations, including alterations in hematopoietic stem cell function, niche composition, and inflammatory signaling. These age-related shifts in the bone marrow microenvironment contribute to dysregulation of immune cell homeostasis and function, impacting neuroinflammatory processes and neuronal health. In our review, we aim to explore the complex cellular and molecular mechanisms that link bone marrow aging to immunosenescence, inflammaging, and neuroinflammation, with a specific focus on their relevance to the pathophysiology of age-related neurological disorders. By exploring this interplay, we strive to provide a comprehensive understanding of how bone marrow aging impacts immune function and contributes to the progression of neurological diseases in aging individuals. Ultimately, this knowledge can hold substantial promise for the development of innovative therapeutic interventions aimed at preserving immune function and mitigating the progression of neurological disorders in the elderly population.
Collapse
Affiliation(s)
- Ludmila Müller
- Max Planck Institute for Human Development, Center for Lifespan Psychology, Berlin, Germany
| | | |
Collapse
|
31
|
Koupourtidou C, Schwarz V, Aliee H, Frerich S, Fischer-Sternjak J, Bocchi R, Simon-Ebert T, Bai X, Sirko S, Kirchhoff F, Dichgans M, Götz M, Theis FJ, Ninkovic J. Shared inflammatory glial cell signature after stab wound injury, revealed by spatial, temporal, and cell-type-specific profiling of the murine cerebral cortex. Nat Commun 2024; 15:2866. [PMID: 38570482 PMCID: PMC10991294 DOI: 10.1038/s41467-024-46625-w] [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: 03/03/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Traumatic brain injury leads to a highly orchestrated immune- and glial cell response partially responsible for long-lasting disability and the development of secondary neurodegenerative diseases. A holistic understanding of the mechanisms controlling the responses of specific cell types and their crosstalk is required to develop an efficient strategy for better regeneration. Here, we combine spatial and single-cell transcriptomics to chart the transcriptomic signature of the injured male murine cerebral cortex, and identify specific states of different glial cells contributing to this signature. Interestingly, distinct glial cells share a large fraction of injury-regulated genes, including inflammatory programs downstream of the innate immune-associated pathways Cxcr3 and Tlr1/2. Systemic manipulation of these pathways decreases the reactivity state of glial cells associated with poor regeneration. The functional relevance of the discovered shared signature of glial cells highlights the importance of our resource enabling comprehensive analysis of early events after brain injury.
Collapse
Affiliation(s)
- Christina Koupourtidou
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Veronika Schwarz
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Hananeh Aliee
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Simon Frerich
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Judith Fischer-Sternjak
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Riccardo Bocchi
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Tatiana Simon-Ebert
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
| | - Swetlana Sirko
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany
- German Centre for Neurodegenerative Diseases, Munich, Germany
| | - Magdalena Götz
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, Munich, Germany
| | - Jovica Ninkovic
- Chair of Cell Biology and Anatomy, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany.
- Munich Cluster for Systems Neurology SYNERGY, LMU Munich, Munich, Germany.
| |
Collapse
|
32
|
Nonaka H, Kondo T, Suga M, Yamanaka R, Sagara Y, Tsukita K, Mitsutomi N, Homma K, Saito R, Miyoshi F, Ohzeki H, Okuyama M, Inoue H. Induced pluripotent stem cell-based assays recapture multiple properties of human astrocytes. J Cell Mol Med 2024; 28:e18214. [PMID: 38509731 PMCID: PMC10955154 DOI: 10.1111/jcmm.18214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
The majority of the population of glial cells in the central nervous system consists of astrocytes, and impairment of astrocytes causes various disorders. It is useful to assess the multiple astrocytic properties in order to understand their complex roles in the pathophysiology. Although we can differentiate human astrocytes from induced pluripotent stem cells (iPSCs), it remains unknown how we can analyse and reveal the multiple properties of astrocytes in complexed human disease conditions. For this purpose, we tested astrocytic differentiation protocols from feeder-free iPSCs based on the previous method with some modifications. Then, we set up extra- and intracellular assessments of iPSC-derived astrocytes by testing cytokine release, calcium influx, autophagy induction and migration. The results led us to analytic methods with conditions in which iPSC-derived astrocytes behave as in vivo. Finally, we applied these methods for modelling an astrocyte-related disease, Alexander disease. An analytic system using iPSC-derived astrocytes could be used to recapture complexities in human astrocyte diseases.
Collapse
Affiliation(s)
- Hideki Nonaka
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Takayuki Kondo
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
- Medical‐risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP)KyotoJapan
| | - Mika Suga
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | - Ryu Yamanaka
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Yukako Sagara
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | - Kayoko Tsukita
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | | | - Kengo Homma
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | - Ryuta Saito
- Mitsubishi Tanabe Pharma CorporationYokohamaJapan
| | | | | | | | - Haruhisa Inoue
- iPSC‐based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC)KyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
- Medical‐risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP)KyotoJapan
| |
Collapse
|
33
|
Nam YR, Kang M, Kim M, Seok MJ, Yang Y, Han YE, Oh SJ, Kim DG, Son H, Chang MY, Lee SH. Preparation of human astrocytes with potent therapeutic functions from human pluripotent stem cells using ventral midbrain patterning. J Adv Res 2024:S2090-1232(24)00112-7. [PMID: 38521186 DOI: 10.1016/j.jare.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/16/2024] [Indexed: 03/25/2024] Open
Abstract
INTRODUCTION Astrocytes are glial-type cells that protect neurons from toxic insults and support neuronal functions and metabolism in a healthy brain. Leveraging these physiological functions, transplantation of astrocytes or their derivatives has emerged as a potential therapeutic approach for neurodegenerative disorders. METHODS To substantiate the clinical application of astrocyte-based therapy, we aimed to prepare human astrocytes with potent therapeutic capacities from human pluripotent stem cells (hPSCs). To that end, we used ventral midbrain patterning during the differentiation of hPSCs into astrocytes, based on the roles of midbrain-specific factors in potentiating glial neurotrophic/anti-inflammatory activity. To assess the therapeutic effects of human midbrain-type astrocytes, we transplanted them into mouse models of Parkinson's disease (PD) and Alzheimer's disease (AD). RESULTS Through a comprehensive series of in-vitro and in-vivo experiments, we were able to establish that the midbrain-type astrocytes exhibited the abilities to effectively combat oxidative stress, counter excitotoxic glutamate, and manage pathological protein aggregates. Our strategy for preparing midbrain-type astrocytes yielded promising results, demonstrating the strong therapeutic potential of these cells in various neurotoxic contexts. Particularly noteworthy is their efficacy in PD and AD-specific proteopathic conditions, in which the midbrain-type astrocytes outperformed forebrain-type astrocytes derived by the same organoid-based method. CONCLUSION The enhanced functions of the midbrain-type astrocytes extended to their ability to release signaling molecules that inhibited neuronal deterioration and senescence while steering microglial cells away from a pro-inflammatory state. This success was evident in both in-vitro studies using human cells and in-vivo experiments conducted in mouse models of PD and AD. In the end, our human midbrain-type astrocytes demonstrated remarkable effectiveness in alleviating neurodegeneration, neuroinflammation, and the pathologies associated with the accumulation of α-synuclein and Amyloid β proteins.
Collapse
Affiliation(s)
- Ye Rim Nam
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Minji Kang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Minji Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Min Jong Seok
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Yunseon Yang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Young Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Korea
| | - Mi-Yoon Chang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Premedicine, College of Medicine, Hanyang University, Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea.
| | - Sang-Hun Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Korea.
| |
Collapse
|
34
|
Mazzitelli M, Ponomareva O, Presto P, John J, Neugebauer V. Impaired amygdala astrocytic signaling worsens neuropathic pain-associated neuronal functions and behaviors. Front Pharmacol 2024; 15:1368634. [PMID: 38576475 PMCID: PMC10991799 DOI: 10.3389/fphar.2024.1368634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction: Pain is a clinically relevant health care issue with limited therapeutic options, creating the need for new and improved analgesic strategies. The amygdala is a limbic brain region critically involved in the regulation of emotional-affective components of pain and in pain modulation. The central nucleus of amygdala (CeA) serves major output functions and receives nociceptive information via the external lateral parabrachial nucleus (PB). While amygdala neuroplasticity has been linked causally to pain behaviors, non-neuronal pain mechanisms in this region remain to be explored. As an essential part of the neuroimmune system, astrocytes that represent about 40-50% of glia cells within the central nervous system, are required for physiological neuronal functions, but their role in the amygdala remains to be determined for pain conditions. In this study, we measured time-specific astrocyte activation in the CeA in a neuropathic pain model (spinal nerve ligation, SNL) and assessed the effects of astrocyte inhibition on amygdala neuroplasticity and pain-like behaviors in the pain condition. Methods and Results: Glial fibrillary acidic protein (GFAP, astrocytic marker) immunoreactivity and mRNA expression were increased at the chronic (4 weeks post-SNL), but not acute (1 week post-SNL), stage of neuropathic pain. In order to determine the contribution of astrocytes to amygdala pain-mechanisms, we used fluorocitric acid (FCA), a selective inhibitor of astrocyte metabolism. Whole-cell patch-clamp recordings were performed from neurons in the laterocapsular division of the CeA (CeLC) obtained from chronic neuropathic rats. Pre-incubation of brain slices with FCA (100 µM, 1 h), increased excitability through altered hyperpolarization-activated current (Ih) functions, without significantly affecting synaptic responses at the PB-CeLC synapse. Intra-CeA injection of FCA (100 µM) had facilitatory effects on mechanical withdrawal thresholds (von Frey and paw pressure tests) and emotional-affective behaviors (evoked vocalizations), but not on facial grimace score and anxiety-like behaviors (open field test), in chronic neuropathic rats. Selective inhibition of astrocytes by FCA was confirmed with immunohistochemical analyses showing decreased astrocytic GFAP, but not NeuN, signal in the CeA. Discussion: Overall, these results suggest a complex modulation of amygdala pain functions by astrocytes and provide evidence for beneficial functions of astrocytes in CeA in chronic neuropathic pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Julia John
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
35
|
Zhou S, Liu C, Wang J, Ye J, Lian Q, Gan L, Deng S, Xu T, Guo Y, Li W, Zhang Z, Yang GY, Tang Y. CCL5 mediated astrocyte-T cell interaction disrupts blood-brain barrier in mice after hemorrhagic stroke. J Cereb Blood Flow Metab 2024; 44:367-383. [PMID: 37974301 PMCID: PMC10870968 DOI: 10.1177/0271678x231214838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
The crosstalk between reactive astrocytes and infiltrated immune cells plays a critical role in maintaining blood-brain barrier (BBB) integrity. However, how astrocytes interact with immune cells and the effect of their interaction on BBB integrity after hemorrhagic stroke are still unclear. By performing RNA sequencing in astrocytes that were activated by interleukin-1α (IL-1α), tumor necrosis factor α (TNFα), and complement component 1q (C1q) treatment, we found CCL5 was among the top upregulated genes. Immunostaining and western blot results demonstrated that CCL5 was increased in mice brain after hemorrhagic stroke. Flow cytometry showed that knockout of astrocytic CCL5 reduced the infiltration of CD8+ but not CD4+ T and myeloid cells into the brain (p < 0.05). In addition, knockout CCL5 in astrocytes increased tight junction-related proteins ZO-1 and Occludin expression; reduced Evans blue leakage, perforin and granzyme B expression; improved neurobehavioral outcomes in hemorrhagic stroke mice (p < 0.05), while transplantation of CD8+ T cells reversed these protective effects. Moreover, co-culture of CD8+ T cells with bEnd.3 cells induced the apoptosis of bEnd.3 cells, which was rescued by inhibiting perforin. In conclusion, our study suggests that CCL5 mediated crosstalk between astrocytes and CD8+ T cells represents an important therapeutic target for protecting BBB in stroke.
Collapse
Affiliation(s)
- Shiyi Zhou
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Liu
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jixian Wang
- Department of Rehabilitation Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Ye
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qianyuan Lian
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Gan
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyu Deng
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tongtong Xu
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyan Guo
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wanlu Li
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Shanghai Sixth People’s Hospital and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Kwok AJ, Lu J, Huang J, Ip BY, Mok VCT, Lai HM, Ko H. High-resolution omics of vascular ageing and inflammatory pathways in neurodegeneration. Semin Cell Dev Biol 2024; 155:30-49. [PMID: 37380595 DOI: 10.1016/j.semcdb.2023.06.005] [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: 04/29/2023] [Accepted: 06/07/2023] [Indexed: 06/30/2023]
Abstract
High-resolution omics, particularly single-cell and spatial transcriptomic profiling, are rapidly enhancing our comprehension of the normal molecular diversity of gliovascular cells, as well as their age-related changes that contribute to neurodegeneration. With more omic profiling studies being conducted, it is becoming increasingly essential to synthesise valuable information from the rapidly accumulating findings. In this review, we present an overview of the molecular features of neurovascular and glial cells that have been recently discovered through omic profiling, with a focus on those that have potentially significant functional implications and/or show cross-species differences between human and mouse, and that are linked to vascular deficits and inflammatory pathways in ageing and neurodegenerative disorders. Additionally, we highlight the translational applications of omic profiling, and discuss omic-based strategies to accelerate biomarker discovery and facilitate disease course-modifying therapeutics development for neurodegenerative conditions.
Collapse
Affiliation(s)
- Andrew J Kwok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jianning Lu
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bonaventure Y Ip
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent C T Mok
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Ming Lai
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Margaret K. L. Cheung Research Centre for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
37
|
Prakash P, Erdjument-Bromage H, O'Dea MR, Munson CN, Labib D, Fossati V, Neubert TA, Liddelow SA. Proteomic profiling of interferon-responsive reactive astrocytes in rodent and human. Glia 2024; 72:625-642. [PMID: 38031883 PMCID: PMC10843807 DOI: 10.1002/glia.24494] [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: 05/16/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Astrocytes are a heterogeneous population of central nervous system glial cells that respond to pathological insults and injury by undergoing a transformation called "reactivity." Reactive astrocytes exhibit distinct and context-dependent cellular, molecular, and functional state changes that can either support or disturb tissue homeostasis. We recently identified a reactive astrocyte sub-state defined by interferon-responsive genes like Igtp, Ifit3, Mx1, and others, called interferon-responsive reactive astrocytes (IRRAs). To further this transcriptomic definition of IRRAs, we wanted to define the proteomic changes that occur in this reactive sub-state. We induced IRRAs in immunopanned rodent astrocytes and human iPSC-differentiated astrocytes using TNF, IL1α, C1Q, and IFNβ and characterized their proteomic profile (both cellular and secreted) using unbiased quantitative proteomics. We identified 2335 unique cellular proteins, including IFIT2/3, IFITM3, OASL1/2, MX1/2/3, and STAT1. We also report that rodent and human IRRAs secrete PAI1, a serine protease inhibitor which may influence reactive states and functions of nearby cells. Finally, we evaluated how IRRAs are distinct from neurotoxic reactive astrocytes (NRAs). While NRAs are described by expression of the complement protein C3, it was not upregulated in IRRAs. Instead, we found ~90 proteins unique to IRRAs not identified in NRAs, including OAS1A, IFIT3, and MX1. Interferon signaling in astrocytes is critical for the antiviral immune response and for regulating synaptic plasticity and glutamate transport mechanisms. How IRRAs contribute to these functions is unknown. This study provides the basis for future experiments to define the functional roles of IRRAs in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Priya Prakash
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Hediye Erdjument-Bromage
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Christy N Munson
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - David Labib
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Thomas A Neubert
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York, USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
38
|
Kouba BR, de Araujo Borba L, Borges de Souza P, Gil-Mohapel J, Rodrigues ALS. Role of Inflammatory Mechanisms in Major Depressive Disorder: From Etiology to Potential Pharmacological Targets. Cells 2024; 13:423. [PMID: 38474387 DOI: 10.3390/cells13050423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The involvement of central and peripheral inflammation in the pathogenesis and prognosis of major depressive disorder (MDD) has been demonstrated. The increase of pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-18, and TNF-α) in individuals with depression may elicit neuroinflammatory processes and peripheral inflammation, mechanisms that, in turn, can contribute to gut microbiota dysbiosis. Together, neuroinflammation and gut dysbiosis induce alterations in tryptophan metabolism, culminating in decreased serotonin synthesis, impairments in neuroplasticity-related mechanisms, and glutamate-mediated excitotoxicity. This review aims to highlight the inflammatory mechanisms (neuroinflammation, peripheral inflammation, and gut dysbiosis) involved in the pathophysiology of MDD and to explore novel anti-inflammatory therapeutic approaches for this psychiatric disturbance. Several lines of evidence have indicated that in addition to antidepressants, physical exercise, probiotics, and nutraceuticals (agmatine, ascorbic acid, and vitamin D) possess anti-inflammatory effects that may contribute to their antidepressant properties. Further studies are necessary to explore the therapeutic benefits of these alternative therapies for MDD.
Collapse
Affiliation(s)
- Bruna R Kouba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Laura de Araujo Borba
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Pedro Borges de Souza
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis 88040-900, SC, Brazil
| |
Collapse
|
39
|
Theus MH. Neuroinflammation and acquired traumatic CNS injury: a mini review. Front Neurol 2024; 15:1334847. [PMID: 38450073 PMCID: PMC10915049 DOI: 10.3389/fneur.2024.1334847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Acquired traumatic central nervous system (CNS) injuries, including traumatic brain injury (TBI) and spinal cord injury (SCI), are devastating conditions with limited treatment options. Neuroinflammation plays a pivotal role in secondary damage, making it a prime target for therapeutic intervention. Emerging therapeutic strategies are designed to modulate the inflammatory response, ultimately promoting neuroprotection and neuroregeneration. The use of anti-inflammatory agents has yielded limited support in improving outcomes in patients, creating a critical need to re-envision novel approaches to both quell deleterious inflammatory processes and upend the progressive cycle of neurotoxic inflammation. This demands a comprehensive exploration of individual, age, and sex differences, including the use of advanced imaging techniques, multi-omic profiling, and the expansion of translational studies from rodents to humans. Moreover, a holistic approach that combines pharmacological intervention with multidisciplinary neurorehabilitation is crucial and must include both acute and long-term care for the physical, cognitive, and emotional aspects of recovery. Ongoing research into neuroinflammatory biomarkers could revolutionize our ability to predict, diagnose, and monitor the inflammatory response in real time, allowing for timely adjustments in treatment regimens and facilitating a more precise evaluation of therapeutic efficacy. The management of neuroinflammation in acquired traumatic CNS injuries necessitates a paradigm shift in our approach that includes combining multiple therapeutic modalities and fostering a more comprehensive understanding of the intricate neuroinflammatory processes at play.
Collapse
Affiliation(s)
- Michelle H. Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, United States
- Center for Engineered Health, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
40
|
Wu Y, Meng X, Cheng WY, Yan Z, Li K, Wang J, Jiang T, Zhou F, Wong KH, Zhong C, Dong Y, Gao S. Can pluripotent/multipotent stem cells reverse Parkinson's disease progression? Front Neurosci 2024; 18:1210447. [PMID: 38356648 PMCID: PMC10864507 DOI: 10.3389/fnins.2024.1210447] [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: 04/22/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by continuous and selective degeneration or death of dopamine neurons in the midbrain, leading to dysfunction of the nigrostriatal neural circuits. Current clinical treatments for PD include drug treatment and surgery, which provide short-term relief of symptoms but are associated with many side effects and cannot reverse the progression of PD. Pluripotent/multipotent stem cells possess a self-renewal capacity and the potential to differentiate into dopaminergic neurons. Transplantation of pluripotent/multipotent stem cells or dopaminergic neurons derived from these cells is a promising strategy for the complete repair of damaged neural circuits in PD. This article reviews and summarizes the current preclinical/clinical treatments for PD, their efficacies, and the advantages/disadvantages of various stem cells, including pluripotent and multipotent stem cells, to provide a detailed overview of how these cells can be applied in the treatment of PD, as well as the challenges and bottlenecks that need to be overcome in future translational studies.
Collapse
Affiliation(s)
- Yongkang Wu
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Xiangtian Meng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wai-Yin Cheng
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zhichao Yan
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keqin Li
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People’s Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Ka-Hing Wong
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
41
|
Stankovic I, Notaras M, Wolujewicz P, Lu T, Lis R, Ross ME, Colak D. Schizophrenia endothelial cells exhibit higher permeability and altered angiogenesis patterns in patient-derived organoids. Transl Psychiatry 2024; 14:53. [PMID: 38263175 PMCID: PMC10806043 DOI: 10.1038/s41398-024-02740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024] Open
Abstract
Schizophrenia (SCZ) is a complex neurodevelopmental disorder characterized by the manifestation of psychiatric symptoms in early adulthood. While many research avenues into the origins of SCZ during brain development have been explored, the contribution of endothelial/vascular dysfunction to the disease remains largely elusive. To model the neuropathology of SCZ during early critical periods of brain development, we utilized patient-derived induced pluripotent stem cells (iPSCs) to generate 3D cerebral organoids and define cell-specific signatures of disease. Single-cell RNA sequencing revealed that while SCZ organoids were similar in their macromolecular diversity to organoids generated from healthy controls (CTRL), SCZ organoids exhibited a higher percentage of endothelial cells when normalized to total cell numbers. Additionally, when compared to CTRL, differential gene expression analysis revealed a significant enrichment in genes that function in vessel formation, vascular regulation, and inflammatory response in SCZ endothelial cells. In line with these findings, data from 23 donors demonstrated that PECAM1+ microvascular vessel-like structures were increased in length and number in SCZ organoids in comparison to CTRL organoids. Furthermore, we report that patient-derived endothelial cells displayed higher paracellular permeability, implicating elevated vascular activity. Collectively, our data identified altered gene expression patterns, vessel-like structural changes, and enhanced permeability of endothelial cells in patient-derived models of SCZ. Hence, brain microvascular cells could play a role in the etiology of SCZ by modulating the permeability of the developing blood brain barrier (BBB).
Collapse
Affiliation(s)
- Isidora Stankovic
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Paul Wolujewicz
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Tyler Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Raphael Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, USA
| | - M Elizabeth Ross
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
42
|
Tsui CT, Mirkiani S, Roszko DA, Churchward MA, Mushahwar VK, Todd KG. In vitro biocompatibility evaluation of functional electrically stimulating microelectrodes on primary glia. Front Bioeng Biotechnol 2024; 12:1351087. [PMID: 38314352 PMCID: PMC10834782 DOI: 10.3389/fbioe.2024.1351087] [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: 12/06/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Neural interfacing devices interact with the central nervous system to alleviate functional deficits arising from disease or injury. This often entails the use of invasive microelectrode implants that elicit inflammatory responses from glial cells and leads to loss of device function. Previous work focused on improving implant biocompatibility by modifying electrode composition; here, we investigated the direct effects of electrical stimulation on glial cells at the electrode interface. A high-throughput in vitro system that assesses primary glial cell response to biphasic stimulation waveforms at 0 mA, 0.15 mA, and 1.5 mA was developed and optimized. Primary mixed glial cell cultures were generated from heterozygous CX3CR-1+/EGFP mice, electrically stimulated for 4 h/day over 3 days using 75 μm platinum-iridium microelectrodes, and biomarker immunofluorescence was measured. Electrodes were then imaged on a scanning electron microscope to assess sustained electrode damage. Fluorescence and electron microscopy analyses suggest varying degrees of localized responses for each biomarker assayed (Hoescht, EGFP, GFAP, and IL-1β), a result that expands on comparable in vivo models. This system allows for the comparison of a breadth of electrical stimulation parameters, and opens another avenue through which neural interfacing device developers can improve biocompatibility and longevity of electrodes in tissue.
Collapse
Affiliation(s)
- Christopher T. Tsui
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - Soroush Mirkiani
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - David A. Roszko
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - Matthew A. Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
- Department of Biological and Environmental Sciences, Concordia University of Edmonton, Edmonton, AB, Canada
| | - Vivian K. Mushahwar
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kathryn G. Todd
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
43
|
Livne-Bar I, Maurya S, Gronert K, Sivak JM. Lipoxins A 4 and B 4 inhibit glial cell activation via CXCR3 signaling in acute retinal neuroinflammation. J Neuroinflammation 2024; 21:18. [PMID: 38212822 PMCID: PMC10782675 DOI: 10.1186/s12974-024-03010-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/01/2024] [Indexed: 01/13/2024] Open
Abstract
Lipoxins are small lipids that are potent endogenous mediators of systemic inflammation resolution in a variety of diseases. We previously reported that Lipoxins A4 and B4 (LXA4 and LXB4) have protective activities against neurodegenerative injury. Yet, lipoxin activities and downstream signaling in neuroinflammatory processes are not well understood. Here, we utilized a model of posterior uveitis induced by lipopolysaccharide endotoxin (LPS), which results in rapid retinal neuroinflammation primarily characterized by activation of resident macroglia (astrocytes and Müller glia), and microglia. Using this model, we observed that each lipoxin reduces acute inner retinal inflammation by affecting endogenous glial responses in a cascading sequence beginning with astrocytes and then microglia, depending on the timing of exposure; prophylactic or therapeutic. Subsequent analyses of retinal cytokines and chemokines revealed inhibition of both CXCL9 (MIG) and CXCL10 (IP10) by each lipoxin, compared to controls, following LPS injection. CXCL9 and CXCL10 are common ligands for the CXCR3 chemokine receptor, which is prominently expressed in inner retinal astrocytes and ganglion cells. We found that CXCR3 inhibition reduces LPS-induced neuroinflammation, while CXCR3 agonism alone induces astrocyte reactivity. Together, these data uncover a novel lipoxin-CXCR3 pathway to promote distinct anti-inflammatory and proresolution cascades in endogenous retinal glia.
Collapse
Affiliation(s)
- Izhar Livne-Bar
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada
| | - Shubham Maurya
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
- Vision Science Program, University of California Berkeley, Berkeley, CA, USA
- Infectious Disease and Immunity Program, University of California Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada.
| |
Collapse
|
44
|
Chen J, Stork T, Kang Y, Nardone KAM, Auer F, Farrell RJ, Jay TR, Heo D, Sheehan A, Paton C, Nagel KI, Schoppik D, Monk KR, Freeman MR. Astrocyte growth is driven by the Tre1/S1pr1 phospholipid-binding G protein-coupled receptor. Neuron 2024; 112:93-112.e10. [PMID: 38096817 PMCID: PMC11073822 DOI: 10.1016/j.neuron.2023.11.008] [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: 01/10/2023] [Revised: 07/31/2023] [Accepted: 11/08/2023] [Indexed: 01/06/2024]
Abstract
Astrocytes play crucial roles in regulating neural circuit function by forming a dense network of synapse-associated membrane specializations, but signaling pathways regulating astrocyte morphogenesis remain poorly defined. Here, we show the Drosophila lipid-binding G protein-coupled receptor (GPCR) Tre1 is required for astrocytes to establish their intricate morphology in vivo. The lipid phosphate phosphatases Wunen/Wunen2 also regulate astrocyte morphology and, via Tre1, mediate astrocyte-astrocyte competition for growth-promoting lipids. Loss of s1pr1, the functional analog of Tre1 in zebrafish, disrupts astrocyte process elaboration, and live imaging and pharmacology demonstrate that S1pr1 balances proper astrocyte process extension/retraction dynamics during growth. Loss of Tre1 in flies or S1pr1 in zebrafish results in defects in simple assays of motor behavior. Tre1 and S1pr1 are thus potent evolutionarily conserved regulators of the elaboration of astrocyte morphological complexity and, ultimately, astrocyte control of behavior.
Collapse
Affiliation(s)
- Jiakun Chen
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Tobias Stork
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yunsik Kang
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Katherine A M Nardone
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Franziska Auer
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ryan J Farrell
- Neuroscience Institute, NYU Medical Center, New York, NY 10016, USA
| | - Taylor R Jay
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dongeun Heo
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy Sheehan
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Cameron Paton
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - David Schoppik
- Departments of Otolaryngology and Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Marc R Freeman
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
45
|
Frumer GR, Shin SH, Jung S, Kim JS. Not just Glia-Dissecting brain macrophages in the mouse. Glia 2024; 72:5-18. [PMID: 37501579 DOI: 10.1002/glia.24445] [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: 04/06/2023] [Revised: 07/05/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
Macrophages have emerged as critical cellular components of the central nervous system (CNS), promoting development, maintenance, and immune defense of the CNS. Here we will review recent advances in our understanding of brain macrophage heterogeneity, including microglia and border-associated macrophages, focusing on the mouse. Emphasis will be given to the discussion of strengths and limitations of the experimental approaches that have led to the recent insights and hold promise to further deepen our mechanistic understanding of brain macrophages that might eventually allow to harness their activities for the management of CNS pathologies.
Collapse
Affiliation(s)
- Gal Ronit Frumer
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sun-Hye Shin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Steffen Jung
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jung-Seok Kim
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
46
|
He J, Liu F, Xu T, Ma J, Yu H, Zhao J, Xie Y, Luo L, Yang Q, Lou T, He L, Sun D. The role of hydrogen therapy in Alzheimer's disease management: Insights into mechanisms, administration routes, and future challenges. Biomed Pharmacother 2023; 168:115807. [PMID: 37913734 DOI: 10.1016/j.biopha.2023.115807] [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: 08/25/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder predominantly affecting the elderly. While conventional pharmacological therapies remain the primary treatment for AD, their efficacy is limited effectiveness and often associated with significant side effects. This underscores the urgent need to explore alternative, non-pharmacological interventions. Oxidative stress has been identified as a central player in AD pathology, influencing various aspects including amyloid-beta metabolism, tau phosphorylation, autophagy, neuroinflammation, mitochondrial dysfunction, and synaptic dysfunction. Among the emerging non-drug approaches, hydrogen therapy has garnered attention for its potential in mitigating these pathological conditions. This review provides a comprehensively overview of the therapeutic potential of hydrogen in AD. We delve into its mechanisms of action, administration routes, and discuss the current challenges and future prospects, with the aim of providing valuable insights to facilitate the clinical application of hydrogen-based therapies in AD management.
Collapse
Affiliation(s)
- Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Fan Liu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Ting Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jing Zhao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yanyan Xie
- The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Li Luo
- Dongguan Hospital, Southern Medical University, Dongguan 523059, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Ting Lou
- Yiwu Center for Disease Control and Prevention, Yiwu 322000, China.
| | - Luqing He
- Department of Science and Education, the Third People's Hospital Health Care Group of Cixi, Ningbo 315300, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
47
|
Wendlandt M, Kürten AJ, Beiersdorfer A, Schubert C, Samad-Yazdtchi K, Sauer J, Pinto MC, Schulz K, Friese MA, Gee CE, Hirnet D, Lohr C. A 2A adenosine receptor-driven cAMP signaling in olfactory bulb astrocytes is unaffected in experimental autoimmune encephalomyelitis. Front Immunol 2023; 14:1273837. [PMID: 38077336 PMCID: PMC10701430 DOI: 10.3389/fimmu.2023.1273837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction The cyclic nucleotide cyclic adenosine monophosphate (cAMP) is a ubiquitous second messenger, which is known to play an important anti-inflammatory role. Astrocytes in the central nervous system (CNS) can modulate inflammation but little is known about the significance of cAMP in their function. Methods We investigated cAMP dynamics in mouse olfactory bulb astrocytes in brain slices prepared from healthy and experimental autoimmune encephalomyelitis (EAE) mice. Results The purinergic receptor ligands adenosine and adenosine triphosphate (ATP) both induced transient increases in cAMP in astrocytes expressing the genetically encoded cAMP sensor Flamindo2. The A2A receptor antagonist ZM241385 inhibited the responses. Similar transient increases in astrocytic cAMP occurred when olfactory receptor neurons were stimulated electrically, resulting in ATP release from the stimulated axons that increased cAMP, again via A2A receptors. Notably, A2A-mediated responses to ATP and adenosine were not different in EAE mice as compared to healthy mice. Discussion Our results indicate that ATP, synaptically released by afferent axons in the olfactory bulb, is degraded to adenosine that acts on A2A receptors in astrocytes, thereby increasing the cytosolic cAMP concentration. However, this pathway is not altered in the olfactory bulb of EAE mice.
Collapse
Affiliation(s)
- Marina Wendlandt
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Alina J. Kürten
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | | | - Charlotte Schubert
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jessica Sauer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - M. Carolina Pinto
- Institute of Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Kristina Schulz
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Manuel A. Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine E. Gee
- Institute of Synaptic Physiology, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Daniela Hirnet
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
48
|
Lavekar SS, Patel MD, Montalvo-Parra MD, Krencik R. Asteroid impact: the potential of astrocytes to modulate human neural networks within organoids. Front Neurosci 2023; 17:1305921. [PMID: 38075269 PMCID: PMC10702564 DOI: 10.3389/fnins.2023.1305921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/08/2023] [Indexed: 02/12/2024] Open
Abstract
Astrocytes are a vital cellular component of the central nervous system that impact neuronal function in both healthy and pathological states. This includes intercellular signals to neurons and non-neuronal cells during development, maturation, and aging that can modulate neural network formation, plasticity, and maintenance. Recently, human pluripotent stem cell-derived neural aggregate cultures, known as neurospheres or organoids, have emerged as improved experimental platforms for basic and pre-clinical neuroscience compared to traditional approaches. Here, we summarize the potential capability of using organoids to further understand the mechanistic role of astrocytes upon neural networks, including the production of extracellular matrix components and reactive signaling cues. Additionally, we discuss the application of organoid models to investigate the astrocyte-dependent aspects of neuropathological diseases and to test astrocyte-inspired technologies. We examine the shortcomings of organoid-based experimental platforms and plausible improvements made possible by cutting-edge neuroengineering technologies. These advancements are expected to enable the development of improved diagnostic strategies and high-throughput translational applications regarding neuroregeneration.
Collapse
Affiliation(s)
| | | | | | - R. Krencik
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
49
|
Kim RD, Marchildon AE, Frazel PW, Hasel P, Guo AX, Liddelow SA. Temporal and spatial analysis of astrocytes following stroke identifies novel drivers of reactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.12.566710. [PMID: 38014211 PMCID: PMC10680590 DOI: 10.1101/2023.11.12.566710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Astrocytes undergo robust gene expression changes in response to a variety of perturbations, including ischemic injury. How these transitions are affected by time, and how heterogeneous and spatially distinct various reactive astrocyte populations are, remain unclear. To address these questions, we performed spatial transcriptomics as well as single nucleus RNAseq of ∼138,000 mouse forebrain astrocytes at 1, 3, and 14 days after ischemic injury. We observed a widespread and temporally diverse response across many astrocyte subtypes. We identified astrocyte clusters unique in injury, including a transiently proliferative substate that may be BRCA1-dependent. We also found an interferon-responsive population that rapidly expands to the perilesion cortex at 1 day and persists up to 14 days post stroke. These lowly abundant, spatially restricted populations are likely functionally important in post-injury stabilization and resolution. These datasets offer valuable insights into injury-induced reactive astrocyte heterogeneity and can be used to guide functional interrogation of biologically meaningful reactive astrocyte substates to understand their pro- and anti-reparative functions following acute injuries such as stroke.
Collapse
|
50
|
Dong S, Liu X, Bi Y, Wang Y, Antony A, Lee D, Huntoon K, Jeong S, Ma Y, Li X, Deng W, Schrank BR, Grippin AJ, Ha J, Kang M, Chang M, Zhao Y, Sun R, Sun X, Yang J, Chen J, Tang SK, Lee LJ, Lee AS, Teng L, Wang S, Teng L, Kim BYS, Yang Z, Jiang W. Adaptive design of mRNA-loaded extracellular vesicles for targeted immunotherapy of cancer. Nat Commun 2023; 14:6610. [PMID: 37857647 PMCID: PMC10587228 DOI: 10.1038/s41467-023-42365-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
The recent success of mRNA therapeutics against pathogenic infections has increased interest in their use for other human diseases including cancer. However, the precise delivery of the genetic cargo to cells and tissues of interest remains challenging. Here, we show an adaptive strategy that enables the docking of different targeting ligands onto the surface of mRNA-loaded small extracellular vesicles (sEVs). This is achieved by using a microfluidic electroporation approach in which a combination of nano- and milli-second pulses produces large amounts of IFN-γ mRNA-loaded sEVs with CD64 overexpressed on their surface. The CD64 molecule serves as an adaptor to dock targeting ligands, such as anti-CD71 and anti-programmed cell death-ligand 1 (PD-L1) antibodies. The resulting immunogenic sEVs (imsEV) preferentially target glioblastoma cells and generate potent antitumour activities in vivo, including against tumours intrinsically resistant to immunotherapy. Together, these results provide an adaptive approach to engineering mRNA-loaded sEVs with targeting functionality and pave the way for their adoption in cancer immunotherapy applications.
Collapse
Affiliation(s)
- Shiyan Dong
- School of Life Science, Jilin University, Changchun, 130012, China
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xuan Liu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Chemical Engineering, Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA, 71272, USA
| | - Ye Bi
- Practice Training Center, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Abin Antony
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - DaeYong Lee
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Seongdong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yifan Ma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Xuefeng Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Weiye Deng
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Benjamin R Schrank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Adam J Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - JongHoon Ha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Minjeong Kang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mengyu Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yarong Zhao
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Rongze Sun
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Xiangshi Sun
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Jie Yang
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Jiayi Chen
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Sarah K Tang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - L James Lee
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, USA
- Spot Biosystems Ltd., Palo Alto, CA, 94305, USA
| | - Andrew S Lee
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518055, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Lirong Teng
- School of Life Science, Jilin University, Changchun, 130012, China
| | - Shengnian Wang
- Chemical Engineering, Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA, 71272, USA.
| | - Lesheng Teng
- School of Life Science, Jilin University, Changchun, 130012, China.
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Zhaogang Yang
- School of Life Science, Jilin University, Changchun, 130012, China.
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|