151
|
Wang X, Chen G, Wan B, Dong Z, Xue Y, Luo Q, Wang D, Lu Y, Zhu L. NRF1-mediated microglial activation triggers high-altitude cerebral edema. J Mol Cell Biol 2022; 14:6608944. [PMID: 35704676 PMCID: PMC9486928 DOI: 10.1093/jmcb/mjac036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/24/2022] [Accepted: 06/13/2022] [Indexed: 12/05/2022] Open
Abstract
High-altitude cerebral edema (HACE) is a potentially fatal encephalopathy associated with a time-dependent exposure to the hypobaric hypoxia of altitude. The formation of HACE is affected by both vasogenic and cytotoxic edema. The over-activated microglia potentiate the damage of blood-brain barrier (BBB) and exacerbate cytotoxic edema. In light with the activation of microglia in HACE, we aimed to investigate whether the over-activated microglia were the key turning point of acute mountain sickness to HACE. In in vivo experiments, by exposing mice to hypobaric hypoxia (7000 m above sea level) to induce HACE model, we found that microglia were activated and migrated to blood vessels. Microglia depletion by PLX5622 obviously relieved brain edema. In in vitro experiments, we found that hypoxia induced cultured microglial activation, leading to the destruction of endothelial tight junction and astrocyte swelling. Up-regulated nuclear respiratory factor 1 (NRF1) accelerated pro-inflammatory factors through transcriptional regulation on nuclear factor kappa B p65 (NF-κB p65) and mitochondrial transcription factor A (TFAM) in activated microglia under hypoxia. NRF1 also up-regulated phagocytosis by transcriptional regulation on caveolin-1 (CAV-1) and adaptor-related protein complex 2 subunit beta (AP2B1). The present study reveals a new mechanism in HACE: hypoxia over-activates microglia through up-regulation of NRF1, which both induces inflammatory response through transcriptionally activating NF-κB p65 and TFAM, and enhances phagocytic function through up-regulation of CAV-1 and AP2B1; hypoxia-activated microglia destroy the integrity of BBB and release pro-inflammatory factors that eventually induce HACE.
Collapse
Affiliation(s)
| | - Guijuan Chen
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Baolan Wan
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Zhangji Dong
- Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong 226019, China
| | - Yan Xue
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Qianqian Luo
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Dan Wang
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Yapeng Lu
- Institute of Special Environmental Medicine, Nantong University, Nantong 226019, China,Co-Innovation Center of Neuroregeneration, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Li Zhu
- Correspondence to: Li Zhu, E-mail:
| |
Collapse
|
152
|
Deng J, Meng F, Zhang K, Gao J, Liu Z, Li M, Liu X, Li J, Wang Y, Zhang L, Tang P. Emerging Roles of Microglia Depletion in the Treatment of Spinal Cord Injury. Cells 2022; 11:cells11121871. [PMID: 35741000 PMCID: PMC9221038 DOI: 10.3390/cells11121871] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Microglia, as the resident immune cells and first responder to neurological insults, play an extremely important role in the pathophysiological process of spinal cord injury. On the one hand, microglia respond rapidly and gather around the lesion in the early stage of injury to exert a protective role, but with the continuous stimulation of the injury, the excessive activated microglia secrete a large number of harmful substances, aggravate the injury of spinal cord tissue, and affect functional recovery. The effects of microglia depletion on the repair of spinal cord injury remain unclear, and there is no uniformly accepted paradigm for the removal methods and timing of microglia depletion, but different microglia depletion strategies greatly affect the outcomes after spinal cord injury. Therefore, this review summarizes the physiological and pathological roles of microglia, especially the effects of microglia depletion on spinal cord injury-sustained microglial depletion would aggravate injury and impair functional recovery, while the short-term depletion of microglial population in diseased conditions seems to improve tissue repair and promote functional improvement after spinal cord injury. Furthermore, we discuss the advantages and disadvantages of major strategies and timing of microglia depletion to provide potential strategy for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Junhao Deng
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Fanqi Meng
- Department of Spine Surgery, Peking University People’s Hospital, Beijing 100044, China;
| | - Kexue Zhang
- Department of Pediatric Surgery, The Chinese PLA General Hospital, Beijing 100853, China;
| | - Jianpeng Gao
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Zhongyang Liu
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Ming Li
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Xiao Liu
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Jiantao Li
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopaedics, The Chinese PLA General Hospital, Beijing 100853, China;
| | - Licheng Zhang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
- Correspondence: (L.Z.); (P.T.)
| | - Peifu Tang
- Senior Department of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China;
- National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing 100853, China; (J.G.); (Z.L.); (M.L.); (X.L.); (J.L.)
- Correspondence: (L.Z.); (P.T.)
| |
Collapse
|
153
|
Liu D, Ji Q, Cheng Y, Liu M, Zhang B, Mei Q, Huan M, Zhou S. Cyclosporine A loaded brain targeting nanoparticle to treat cerebral ischemia/reperfusion injury in mice. J Nanobiotechnology 2022; 20:256. [PMID: 35658867 PMCID: PMC9164331 DOI: 10.1186/s12951-022-01474-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/23/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Ischemic stroke is one of the main causes of death and disability in the world. The treatment for ischemic stroke is to restore blood perfusion as soon as possible. However, when ischemic brain tissue is re-perfused by blood, the mitochondrial permeability transition pore (mPTP) in neuron and microglia is excessively opened, resulting in the apoptosis of neuron and nerve inflammation. This aggravates nerve injury. Cyclosporine A (CsA) inhibits the over-opening of mPTP, subsequently reducing the release of ROS and the apoptosis of cerebral ischemia/reperfusion injured neuron and microglia. However, CsA is insoluble in water and present in high concentrations in lymphatic tissue. Herein, cerebral infarction tissue targeted nanoparticle (CsA@HFn) was developed to treat cerebral ischemia/reperfusion injury. RESULTS CsA@HFn efficiently penetrated the blood-brain barrier (BBB) and selectively accumulated in ischemic area, inhibiting the opening of mPTP and ROS production in neuron. This subsequently reduced the apoptosis of neuron and the damage of BBB. Consequently, CsA@HFn significantly reduced the infarct area. Moreover, CsA@HFn inhibited the recruitment of astrocytes and microglia in ischemic region and polarized microglia into M2 type microglia, which subsequently alleviated the nerve inflammation. CONCLUSIONS CsA@HFn showed a significant therapeutic effect on cerebral ischemia/reperfusion injury by alleviating the apoptosis of neuron, nerve inflammation and the damage of BBB in ischemic area. CsA@HFn has great potential in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Daozhou Liu
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Qifeng Ji
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Ying Cheng
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Miao Liu
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Bangle Zhang
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Qibing Mei
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Menglei Huan
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| | - Siyuan Zhou
- grid.233520.50000 0004 1761 4404Department of Pharmaceutics, School of Pharmacy, Air Force Medical University, Changle West Road 169, Xi’an, 710032 Shaanxi China
| |
Collapse
|
154
|
Kwon V, Cai P, Dixon CT, Hamlin V, Spencer CG, Rojas AM, Hamilton M, Shiau CE. Peripheral NOD-like receptor deficient inflammatory macrophages trigger neutrophil infiltration into the brain disrupting daytime locomotion. Commun Biol 2022; 5:464. [PMID: 35577844 PMCID: PMC9110401 DOI: 10.1038/s42003-022-03410-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/25/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammation is known to disrupt normal behavior, yet the underlying neuroimmune interactions remain elusive. Here, we investigated whether inappropriate macrophage-evoked inflammation alters CNS control of daily-life animal locomotion using a set of zebrafish mutants selected for specific macrophage dysfunction and microglia deficiency. Large-scale genetic and computational analyses revealed that NOD-like receptor nlrc3l mutants are capable of normal motility and visuomotor response, but preferentially swim less in the daytime, suggesting possible low motivation rather than physical impairment. Examining their brain activities and structures implicates impaired dopaminergic descending circuits, where neutrophils abnormally infiltrate. Furthermore, neutrophil depletion recovered daytime locomotion. Restoring wild-type macrophages reversed behavioral and neutrophil aberrations, while three other microglia-lacking mutants failed to phenocopy nlrc3l mutants. Overall, we reveal how peripheral inflammatory macrophages with elevated pro-inflammatory cues (including il1β, tnfα, cxcl8a) in the absence of microglia co-opt neutrophils to infiltrate the brain, thereby potentially enabling local circuitry modulation affecting daytime locomotion.
Collapse
Affiliation(s)
- Victoria Kwon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Peiwen Cai
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cameron T Dixon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Victoria Hamlin
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Caroline G Spencer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alison M Rojas
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew Hamilton
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Celia E Shiau
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
155
|
Yan Y, Liu Y, Yang Y, Ding Y, Sun X. Carnosol suppresses microglia cell inflammation and apoptosis through PI3K/AKT/mTOR signaling pathway. Immunopharmacol Immunotoxicol 2022; 44:656-662. [PMID: 35521965 DOI: 10.1080/08923973.2022.2074448] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Ischemic stroke is the most common type of acute cerebrovascular disease. Carnosol is a polyphenol compound extracted from rosemary. This study aimed to explore the effects of carnosol on the oxygen-glucose deprivation (OGD) treated BV2 microglia cells. MTT and EdU assays were used to detect the cell viability and proliferation. Flow cytometry was conducted to measure the apoptosis rates. Additionally, the protein expression was determined by western blot. The inflammatory factors and antioxidant indexes were detected by corresponding kits. Carnosol promoted the proliferation and inhibited the apoptosis of the OGD treated BV2 cells. What's more, the protein expressions of PCNA and Bcl-2 were up-regulated, the Bax expression was down-regulated after carnosol treatment. In addition, carnosol decreased the levels of MDA, LPO, TNF-α, IL-1β and IL-6, and increased the levels of GSH, SOD, IL-4 and IL-10 in the OGD treated BV2 cells. Furthermore, the PI3K/AKT/mTOR signaling pathway was activated after carnosol treatment and inhibition of the PI3K/AKT/mTOR signaling pathway reversed the carnosol effects. Carnosol promotes the proliferation, inhibits the apoptosis, relieves the oxidative damage and inflammation of OGD treated cells through regulating the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuhan Yan
- Department of Geriatrics, General Hospital of Eastern Theater Command, Nanjing, Jiangsu 210002, China
| | - Yu Liu
- Department of Geriatrics, General Hospital of Eastern Theater Command, Nanjing, Jiangsu 210002, China
| | - Yujiao Yang
- Department of Geriatrics, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213000, China
| | - Yi Ding
- Department of Geriatrics, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213000, China
| | - Xin Sun
- Department of Geriatrics, The First People's Hospital of Changzhou, Changzhou, Jiangsu 213000, China
| |
Collapse
|
156
|
Wang J, Xu L, Lin W, Yao Y, Li H, Shen G, Cao X, He N, Chen J, Hu J, Zheng M, Song X, Ding Y, Shen Y, Zhong J, Wang LL, Chen YY, Zhu Y. Single-cell transcriptome analysis reveals the immune heterogeneity and the repopulation of microglia by Hif1α in mice after spinal cord injury. Cell Death Dis 2022; 13:432. [PMID: 35504882 PMCID: PMC9065023 DOI: 10.1038/s41419-022-04864-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is regarded as a vital pathological process in spinal cord injury (SCI), which removes damaged tissue, secretes cytokines, and facilitates regeneration. Repopulation of microglia has been shown to favor recovery from SCI. However, the origin and regulatory factors of microglia repopulation after SCI remain unknown. Here, we used single-cell RNA sequencing to portray the dynamic transcriptional landscape of immune cells during the early and late phases of SCI in mice. B cells and migDCs, located in the meninges under physiological conditions, are involved in immune surveillance. Microglia quickly reduced, and peripheral myeloid cells infiltrated three days-post-injury (dpi). At 14 dpi, microglia repopulated, myeloid cells were reduced, and lymphocytes infiltrated. Importantly, genetic lineage tracing of nestin+ and Cx3cr1+ cells in vivo showed that the repopulation of microglia was derived from residual microglia after SCI. We found that residual microglia regress to a developmental growth state in the early stages after SCI. Hif1α promotes microglial proliferation. Conditional ablation of Hif1α in microglia causes larger lesion sizes, fewer axon fibers, and impaired functional recovery in the late stages after SCI. Our results mapped the immune heterogeneity in SCI and raised the possibility that targeting Hif1α may help in axon regeneration and functional recovery after SCI.
Collapse
Affiliation(s)
- Jingyu Wang
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Lintao Xu
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Weiwei Lin
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Yin Yao
- grid.412465.0Department of Neurointensive Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Heyangzi Li
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Gerong Shen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Cao
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning He
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Chen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Hu
- grid.506977.a0000 0004 1757 7957School of Basic Medical Sciences & Forensic Medicine of Hangzhou Medical College, Hangzhou, China
| | - Mingzhi Zheng
- grid.506977.a0000 0004 1757 7957School of Basic Medical Sciences & Forensic Medicine of Hangzhou Medical College, Hangzhou, China
| | - Xinghui Song
- grid.13402.340000 0004 1759 700XCore Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuemin Ding
- grid.13402.340000 0004 1759 700XSchool of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yueliang Shen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinjie Zhong
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin-lin Wang
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying-ying Chen
- grid.13402.340000 0004 1759 700XDepartment of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongjian Zhu
- grid.412465.0Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, China ,Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
157
|
Cai Y, Liu J, Wang B, Sun M, Yang H. Microglia in the Neuroinflammatory Pathogenesis of Alzheimer's Disease and Related Therapeutic Targets. Front Immunol 2022; 13:856376. [PMID: 35558075 PMCID: PMC9086828 DOI: 10.3389/fimmu.2022.856376] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, characterized by progressive neuron degeneration or loss due to excessive accumulation of β-amyloid (Aβ) peptides, formation of neurofibrillary tangles (NFTs), and hyperphosphorylated tau. The treatment of AD has been only partially successful as the majority of the pharmacotherapies on the market may alleviate some of the symptoms. In the occurrence of AD, increasing attention has been paid to neurodegeneration, while the resident glial cells, like microglia are also observed. Microglia, a kind of crucial glial cells associated with the innate immune response, functions as double-edge sword role in CNS. They exert a beneficial or detrimental influence on the adjacent neurons through secretion of both pro-inflammatory cytokines as well as neurotrophic factors. In addition, their endocytosis of debris and toxic protein like Aβ and tau ensures homeostasis of the neuronal microenvironment. In this review, we will systematically summarize recent research regarding the roles of microglia in AD pathology and latest microglia-associated therapeutic targets mainly including pro-inflammatory genes, anti-inflammatory genes and phagocytosis at length, some of which are contradictory and controversial and warrant to further be investigated.
Collapse
Affiliation(s)
| | | | | | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Yang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
158
|
Cai L, Gong Q, Qi L, Xu T, Suo Q, Li X, Wang W, Jing Y, Yang D, Xu Z, Yuan F, Tang Y, Yang G, Ding J, Chen H, Tian H. ACT001 attenuates microglia-mediated neuroinflammation after traumatic brain injury via inhibiting AKT/NFκB/NLRP3 pathway. Cell Commun Signal 2022; 20:56. [PMID: 35461293 PMCID: PMC9035258 DOI: 10.1186/s12964-022-00862-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/12/2022] [Indexed: 12/20/2022] Open
Abstract
Abstract
Background
Microglia-mediated neuroinflammatory response following traumatic brain injury (TBI) is considered as a vital secondary injury factor, which drives trauma-induced neurodegeneration and is lack of efficient treatment. ACT001, a sesquiterpene lactone derivative, is reportedly involved in alleviation of inflammatory response. However, little is known regarding its function in regulating innate immune response of central nervous system (CNS) after TBI. This study aimed to investigate the role and underlying mechanism of ACT001 in TBI.
Methods
Controlled cortical impact (CCI) models were used to establish model of TBI. Cresyl violet staining, evans blue extravasation, neurobehavioral function assessments, immunofluorescence and transmission electron microscopy were used to evaluate therapeutic effects of ACT001 in vivo. Microglial depletion was induced by administering mice with colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622. Cell-cell interaction models were established as co-culture system to simulate TBI conditions in vitro. Cytotoxic effect of ACT001 on cell viability was assessed by cell counting kit-8 and activation of microglia cells were induced by Lipopolysaccharides (LPS). Pro-inflammatory cytokines expression was determined by Real-time PCR and nitric oxide production. Apoptotic cells were detected by TUNEL and flow cytometry assays. Tube formation was performed to evaluate cellular angiogenic ability. ELISA and western blot experiments were used to determine proteins expression. Pull-down assay was used to analyze proteins that bound ACT001.
Results
ACT001 relieved the extent of blood-brain barrier integrity damage and alleviated motor function deficits after TBI via reducing trauma-induced activation of microglia cells. Delayed depletion of microglia with PLX5622 hindered therapeutic effect of ACT001. Furthermore, ACT001 alleviated LPS-induced activation in mouse and rat primary microglia cells. Besides, ACT001 was effective in suppressing LPS-induced pro-inflammatory cytokines production in BV2 cells, resulting in reduction of neuronal apoptosis in HT22 cells and improvement of tube formation in bEnd.3 cells. Mechanism by which ACT001 functioned was related to AKT/NFκB/NLRP3 pathway. ACT001 restrained NFκB nuclear translocation in microglia cells through inhibiting AKT phosphorylation, resulting in decrease of NLRP3 inflammasome activation, and finally down-regulated microglial neuroinflammatory response.
Conclusions
Our study indicated that ACT001 played critical role in microglia-mediated neuroinflammatory response and might be a novel potential chemotherapeutic drug for TBI.
Collapse
|
159
|
Zheng RZ, Lee KY, Qi ZX, Wang Z, Xu ZY, Wu XH, Mao Y. Neuroinflammation Following Traumatic Brain Injury: Take It Seriously or Not. Front Immunol 2022; 13:855701. [PMID: 35392083 PMCID: PMC8981520 DOI: 10.3389/fimmu.2022.855701] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/23/2022] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with high mortality and disability, with a substantial socioeconomic burden. With the standardization of the treatment process, there is increasing interest in the role that the secondary insult of TBI plays in outcome heterogeneity. The secondary insult is neither detrimental nor beneficial in an absolute sense, among which the inflammatory response was a complex cascade of events and can thus be regarded as a double-edged sword. Therefore, clinicians should take the generation and balance of neuroinflammation following TBI seriously. In this review, we summarize the current human and animal model studies of neuroinflammation and provide a better understanding of the inflammatory response in the different stages of TBI. In particular, advances in neuroinflammation using proteomic and transcriptomic techniques have enabled us to identify a functional specific delineation of the immune cell in TBI patients. Based on recent advances in our understanding of immune cell activation, we present the difference between diffuse axonal injury and focal brain injury. In addition, we give a figurative profiling of the general paradigm in the pre- and post-injury inflammatory settings employing a bow-tie framework.
Collapse
Affiliation(s)
- Rui-Zhe Zheng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Kuin-Yu Lee
- Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zeng-Xin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ze-Yu Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xue-Hai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
160
|
Distinct phases of adult microglia proliferation: a Myc-mediated early phase and a Tnfaip3-mediated late phase. Cell Discov 2022; 8:34. [PMID: 35411038 PMCID: PMC9001707 DOI: 10.1038/s41421-022-00377-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
Microgliosis is a hallmark of many neurological diseases, including Alzheimer’s disease, stroke, seizure, traumatic brain and spinal cord injuries, and peripheral and optic nerve injuries. Recent studies have shown that the newly self-renewed microglia have specific neurological functions. However, the mechanism of adult microglia proliferation remains largely unclear. Here, with single-cell RNA sequencing, flow cytometry, and immunohistochemistry, we demonstrate that the sciatic nerve injury induced two distinct phases of microglia proliferation in mouse spinal cord, each with different gene expression profiles. We demonstrate that the transcription factor Myc was transiently upregulated in spinal cord microglia after nerve injury to mediate an early phase microglia proliferation. On the other hand, we reveal that the tumor-necrosis factor alpha-induced protein 3 (Tnfaip3) was downregulated to mediate the Myc-independent late-phase microglia proliferation. We show that cyclin dependent kinase 1, a kinase with important function in the M phase of the cell cycle, was involved only in the early phase. We reveal that although the early phase was neither necessary nor sufficient for the late phase proliferation, the late-phase suppressed the early phase microglia proliferation in the spinal cord. Finally, we demonstrate that the termination of spinal cord microglia proliferation required both Myc and Tnfaip3 to resume their baseline expression. Thus, we have delineated an interactive signaling network in the proliferation of differentiated microglia.
Collapse
|
161
|
Du Y, Brennan FH, Popovich PG, Zhou M. Microglia maintain the normal structure and function of the hippocampal astrocyte network. Glia 2022; 70:1359-1379. [PMID: 35394085 PMCID: PMC9324808 DOI: 10.1002/glia.24179] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 12/14/2022]
Abstract
Microglial control of activity‐dependent plasticity and synaptic remodeling in neuronal networks has been the subject of intense research in the past several years. Although microglia–neuron interactions have been extensively studied, less is known about how microglia influence astrocyte‐dependent control over neuronal structure and function. Here, we explored a role for microglia in regulating the structure and function of the astrocyte syncytium in mouse hippocampus. After depleting microglia using a CSF1R antagonist (PLX5622, Plexxikon), we observed severe disruption of astrocyte syncytial isopotentiality and dye coupling. A decrease in astrocyte‐specific gap junction connexin (Cx) 30 and 43 expression, at least partially accounts for these microglia‐dependent changes in astrocytes. Because neuronal function requires intact astrocyte coupling, we also evaluated the effects of microglia depletion on synaptic transmission in the hippocampus. Without microglia, the strength of synaptic transmission was reduced at baseline and after long‐term potentiation (LTP). Conversely, priming microglia with systemic injections of lipopolysaccharide enhanced CA3‐CA1 synaptic transmission. This microglia‐induced scaling of synaptic transmission was associated with increased expression of post‐synaptic scaffold proteins (Homer1) in CA1. However, astrocyte network function was not affected by microglia priming, indicating that microglia‐dependent effects on astrocytes and neurons vary across functional states. Through manipulation of microglia in the brain, our results reveal the importance of microglia in homeostatic regulation of the astrocyte syncytium and scaling of synaptic transmission. These novel mechanisms uncover a new direction for future studies interrogating microglia function in various physiological and pathological contexts.
Collapse
Affiliation(s)
- Yixing Du
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Min Zhou
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
162
|
Liu D, Lv F, Min S, Yang Y, Chen L. Inhibition of NLRP3 inflammasome-mediated neuroinflammation alleviates electroconvulsive shock-induced memory impairment via regulation of hippocampal synaptic plasticity in depressive rats. Behav Brain Res 2022; 428:113879. [PMID: 35390431 DOI: 10.1016/j.bbr.2022.113879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022]
Abstract
Electroconvulsive shock has been considered one of the most effective treatment modalities for major depressive disorder. The association of acute transitory neuroinflammation in the hippocampus following electroconvulsive therapy with transient learning and memory impairment limits its clinical application. Whereas the NLRP3 inflammatory pathway is deemed to serve a key role in neuroinflammatory regulation, we aimed to examine if NLRP3 inflammasome activation was linked to electroconvulsive shock (ECS)-induced neuroinflammation and cognitive deficits. The depressed rats were modeled with chronic unpredictable mild stress. Their depression-like behaviors and cognitive performance were evaluated via sucrose preference test, forced swim test, open field test, and Morris water maze test. The NLRP3 expression was determined by western blot. The hippocampal CA1 region was immunohistochemically and electron-microscopically examined, respectively, for the activation of Iba-1 positive microglia and the ultrastructure of synapses. In this work, we found that ECS induced microglial activation in the rat hippocampal CA1 region. Pharmacological inhibition of NLRP3 inflammasome with MCC950 (NLRP3 inhibitor) in vivo significantly alleviated ECS-induced spatial learning and memory impairment, partially reversed neuroinflammation, and synaptic structural plasticity in the damaged hippocampal CA1 region, and reduced synapse associated protein expression and microglial activation. It offers a potential new approach for the prevention and treatment of cognitive decline following electroconvulsive therapy.
Collapse
Affiliation(s)
- Di Liu
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Lv
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - You Yang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lihao Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
163
|
Han J, Chitu V, Stanley ER, Wszolek ZK, Karrenbauer VD, Harris RA. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci 2022; 79:219. [PMID: 35366105 PMCID: PMC8976111 DOI: 10.1007/s00018-022-04225-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson's disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | | | - Virginija Danylaité Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine L8:04, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
164
|
García-Juárez M, Camacho-Morales A. Defining the role of anti- and pro-inflammatory outcomes of Interleukin-6 in mental health. Neuroscience 2022; 492:32-46. [DOI: 10.1016/j.neuroscience.2022.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023]
|
165
|
Folick A, Cheang RT, Valdearcos M, Koliwad SK. Metabolic factors in the regulation of hypothalamic innate immune responses in obesity. Exp Mol Med 2022; 54:393-402. [PMID: 35474339 PMCID: PMC9076660 DOI: 10.1038/s12276-021-00666-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022] Open
Abstract
The hypothalamus is a central regulator of body weight and energy homeostasis. There is increasing evidence that innate immune activation in the mediobasal hypothalamus (MBH) is a key element in the pathogenesis of diet-induced obesity. Microglia, the resident immune cells in the brain parenchyma, have been shown to play roles in diverse aspects of brain function, including circuit refinement and synaptic pruning. As such, microglia have also been implicated in the development and progression of neurological diseases. Microglia express receptors for and are responsive to a wide variety of nutritional, hormonal, and immunological signals that modulate their distinct functions across different brain regions. We showed that microglia within the MBH sense and respond to a high-fat diet and regulate the function of hypothalamic neurons to promote food intake and obesity. Neurons, glia, and immune cells within the MBH are positioned to sense and respond to circulating signals that regulate their capacity to coordinate aspects of systemic energy metabolism. Here, we review the current knowledge of how these peripheral signals modulate the innate immune response in the MBH and enable microglia to regulate metabolic control.
Collapse
Affiliation(s)
- Andrew Folick
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rachel T Cheang
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA
| | - Martin Valdearcos
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| | - Suneil K Koliwad
- Diabetes Center and Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
166
|
Osorio D, Zhong Y, Li G, Xu Q, Yang Y, Tian Y, Chapkin RS, Huang JZ, Cai JJ. scTenifoldKnk: An efficient virtual knockout tool for gene function predictions via single-cell gene regulatory network perturbation. PATTERNS (NEW YORK, N.Y.) 2022; 3:100434. [PMID: 35510185 PMCID: PMC9058914 DOI: 10.1016/j.patter.2022.100434] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/13/2021] [Accepted: 01/04/2022] [Indexed: 11/20/2022]
Abstract
Gene knockout (KO) experiments are a proven, powerful approach for studying gene function. However, systematic KO experiments targeting a large number of genes are usually prohibitive due to the limit of experimental and animal resources. Here, we present scTenifoldKnk, an efficient virtual KO tool that enables systematic KO investigation of gene function using data from single-cell RNA sequencing (scRNA-seq). In scTenifoldKnk analysis, a gene regulatory network (GRN) is first constructed from scRNA-seq data of wild-type samples, and a target gene is then virtually deleted from the constructed GRN. Manifold alignment is used to align the resulting reduced GRN to the original GRN to identify differentially regulated genes, which are used to infer target gene functions in analyzed cells. We demonstrate that the scTenifoldKnk-based virtual KO analysis recapitulates the main findings of real-animal KO experiments and recovers the expected functions of genes in relevant cell types.
Collapse
Affiliation(s)
- Daniel Osorio
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Yan Zhong
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai 200062, China
| | - Guanxun Li
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Yongjian Yang
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Robert S. Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Jianhua Z. Huang
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
- School of Data Science, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
- Interdisciplinary Program of Genetics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
167
|
Qu Y, Wang L, Mao Y. Gallic acid attenuates cerebral ischemia/re-perfusion-induced blood-brain barrier injury by modifying polarization of microglia. J Immunotoxicol 2022; 19:17-26. [PMID: 35254962 DOI: 10.1080/1547691x.2022.2043494] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Microglia, the main immune effector cells in the central nervous system, play a dual role in the function/structure of the blood-brain barrier (BBB) and brain health. During and soon after a cerebral ischemic injury, microglia produce neurotrophic factors and neurotoxins that can impact on the injury itself and pathology progression. At the same time, microglia undergo polarization to M1 or M2 pro- vs. anti-inflammatory subtypes that also help drive the outcome of the injury process. Thus, agents that can mitigate cerebral ischemic injury progression, promote protective functions of microglia, and help maintain BBB and overall brain health/host neurologic function after a cerebral ischemic event would be of great use in clinical settings. Protective effects from gallic acid (GA) in cerebral ischemia/re-perfusion-induced injury to the BBB and other sites in the brain have not yet been assessed. To address this, a middle cerebral artery occlusion (MCAO) method was used to establish an experimental ischemic stroke model in mice. Mice were placed in sham operation (Sham), model (MCAO), MCAO + GA (50 mg/kg), MCAO + GA (100 mg/kg), or MCAO + GA (150 mg/kg) groups. At various times post-stroke, cerebral infarct volume and host neurological function were evaluated. In addition, qRT-PCR, Western blotting, and ELISA were used to evaluate the expression and tissue content of microglia-related factors. The results showed GA treatment protected the integrity of the BBB, significantly reduced brain edema, and helped lead to improved neurological function scores in the MCAO mice. Whether these changes were due to that GA attenuated cerebral ischemia/re-perfusion-induced activation of microglial cells overall, in part, by inhibiting their polarization to the M1 subtype, is uncertain. Taking these outcomes together, for now it is reasonable to suggest that use of GA either as a prophylactic or immediately in the event of a cerebral ischemic event/stroke could help to promote neuronal survival and allow for a more likely of host neurological function over time.
Collapse
Affiliation(s)
- Yang Qu
- Neurology Department, Liaocheng People's Hospital, Liaocheng, China
| | - Lin Wang
- Neurology Department, Liaocheng People's Hospital, Liaocheng, China
| | - Yanfang Mao
- Neurology Department, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
168
|
Adams RC, Carter-Cusack D, Shaikh SN, Llanes GT, Johnston RL, Quaife-Ryan G, Boyle G, Koufariotis LT, Möller A, Blazar BR, Vukovic J, MacDonald KPA. Donor bone marrow-derived macrophage MHC II drives neuroinflammation and altered behavior during chronic GVHD in mice. Blood 2022; 139:1389-1408. [PMID: 34570880 PMCID: PMC8900272 DOI: 10.1182/blood.2021011671] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/03/2021] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains the leading cause of nonrelapse mortality after allogeneic stem cell transplantation for hematological malignancies. Manifestations of GVHD in the central nervous system (CNS) present as neurocognitive dysfunction in up to 60% of patients; however, the mechanisms driving chronic GVHD (cGVHD) in the CNS are yet to be elucidated. Our studies of murine cGVHD revealed behavioral deficits associated with broad neuroinflammation and persistent Ifng upregulation. By flow cytometry, we observed a proportional shift in the donor-derived T-cell population in the cGVHD brain from early CD8 dominance to later CD4 sequestration. RNA sequencing of the hippocampus identified perturbations to structural and functional synapse-related gene expression, together with the upregulation of genes associated with interferon-γ responses and antigen presentation. Neuroinflammation in the cortex of mice and humans during acute GVHD was recently shown to be mediated by resident microglia-derived tumor necrosis factor. In contrast, infiltration of proinflammatory major histocompatibility complex (MHC) class II+ donor bone marrow (BM)-derived macrophages (BMDMs) was identified as a distinguishing feature of CNS cGVHD. Donor BMDMs, which composed up to 50% of the CNS myeloid population, exhibited a transcriptional signature distinct from resident microglia. Recipients of MHC class II knockout BM grafts exhibited attenuated neuroinflammation and behavior comparable to controls, suggestive of a critical role of donor BMDM MHC class II expression in CNS cGVHD. Our identification of disease mediators distinct from those in the acute phase indicates the necessity to pursue alternative therapeutic targets for late-stage neurological manifestations.
Collapse
Affiliation(s)
- Rachael C Adams
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Dylan Carter-Cusack
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Samreen N Shaikh
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Genesis T Llanes
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rebecca L Johnston
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gregory Quaife-Ryan
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Glen Boyle
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lambros T Koufariotis
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Möller
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bruce R Blazar
- Masonic Cancer Center and
- Division of Blood & Marrow Transplant & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN; and
| | - Jana Vukovic
- Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, Australia
| | - Kelli P A MacDonald
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
169
|
Temporal and spatial cellular and molecular pathological alterations with single-cell resolution in the adult spinal cord after injury. Signal Transduct Target Ther 2022; 7:65. [PMID: 35232960 PMCID: PMC8888618 DOI: 10.1038/s41392-022-00885-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/04/2022] [Accepted: 01/09/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) involves diverse injury responses in different cell types in a temporally and spatially specific manner. Here, using single-cell transcriptomic analyses combined with classic anatomical, behavioral, electrophysiological analyses, we report, with single-cell resolution, temporal molecular and cellular changes in crush-injured adult mouse spinal cord. Data revealed pathological changes of 12 different major cell types, three of which infiltrated into the spinal cord at distinct times post-injury. We discovered novel microglia and astrocyte subtypes in the uninjured spinal cord, and their dynamic conversions into additional stage-specific subtypes/states. Most dynamic changes occur at 3-days post-injury and by day-14 the second wave of microglial activation emerged, accompanied with changes in various cell types including neurons, indicative of the second round of attacks. By day-38, major cell types are still substantially deviated from uninjured states, demonstrating prolonged alterations. This study provides a comprehensive mapping of cellular/molecular pathological changes along the temporal axis after SCI, which may facilitate the development of novel therapeutic strategies, including those targeting microglia.
Collapse
|
170
|
Leiter O, Zhuo Z, Rust R, Wasielewska JM, Grönnert L, Kowal S, Overall RW, Adusumilli VS, Blackmore DG, Southon A, Ganio K, McDevitt CA, Rund N, Brici D, Mudiyan IA, Sykes AM, Rünker AE, Zocher S, Ayton S, Bush AI, Bartlett PF, Hou ST, Kempermann G, Walker TL. Selenium mediates exercise-induced adult neurogenesis and reverses learning deficits induced by hippocampal injury and aging. Cell Metab 2022; 34:408-423.e8. [PMID: 35120590 DOI: 10.1016/j.cmet.2022.01.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/01/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022]
Abstract
Although the neurogenesis-enhancing effects of exercise have been extensively studied, the molecular mechanisms underlying this response remain unclear. Here, we propose that this is mediated by the exercise-induced systemic release of the antioxidant selenium transport protein, selenoprotein P (SEPP1). Using knockout mouse models, we confirmed that SEPP1 and its receptor low-density lipoprotein receptor-related protein 8 (LRP8) are required for the exercise-induced increase in adult hippocampal neurogenesis. In vivo selenium infusion increased hippocampal neural precursor cell (NPC) proliferation and adult neurogenesis. Mimicking the effect of exercise through dietary selenium supplementation restored neurogenesis and reversed the cognitive decline associated with aging and hippocampal injury, suggesting potential therapeutic relevance. These results provide a molecular mechanism linking exercise-induced changes in the systemic environment to the activation of quiescent hippocampal NPCs and their subsequent recruitment into the neurogenic trajectory.
Collapse
Affiliation(s)
- Odette Leiter
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Zhan Zhuo
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Ruslan Rust
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Joanna M Wasielewska
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lisa Grönnert
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Susann Kowal
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Rupert W Overall
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Vijay S Adusumilli
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Daniel G Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Adam Southon
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Katherine Ganio
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Christopher A McDevitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Nicole Rund
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - David Brici
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | | | - Alexander M Sykes
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Annette E Rünker
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Sara Zocher
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Scott Ayton
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Ashley I Bush
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Biology, Southern University of Science and Technology, Shenzhen, China.
| | - Gerd Kempermann
- CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| | - Tara L Walker
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| |
Collapse
|
171
|
Ahn K, Lee SJ, Mook-Jung I. White matter-associated microglia: New players in brain aging and neurodegenerative diseases. Ageing Res Rev 2022; 75:101574. [PMID: 35093614 DOI: 10.1016/j.arr.2022.101574] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
There has been growing interest in brain aging and rejuvenation. It is well known that brain aging is one of the leading causes of neurodegenerative diseases, such as Alzheimer's disease, but brain aging alone can cause cognitive decline. Microglia are thought to act as 'conductors' of white matter aging by modulating diverse glial cells and phagocytosing white matter-derived myelin debris. A recent study identified a specific subpopulation of microglia in the white matter of aged mice, termed white matter-associated microglia (WAM). Additionally, senescent microglia show impaired phagocytic function and altered lipid metabolism, which cause accumulation of lipid metabolites and eventually lead to myelin sheath degeneration. These results suggest that senescent WAM could be pivotal players in axonal loss during brain aging. The aim of this review is to assess the current state of knowledge on brain aging, with an emphasis on the roles of the white matter and microglia, and suggest potential approaches for rejuvenating the aged brain.
Collapse
Affiliation(s)
- Kyusik Ahn
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
172
|
Li Q, Wu Y, Chen J, Xuan A, Wang X. Microglia and immunotherapy in Alzheimer's disease. Acta Neurol Scand 2022; 145:273-278. [PMID: 34779509 DOI: 10.1111/ane.13551] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/21/2021] [Accepted: 10/31/2021] [Indexed: 01/01/2023]
Abstract
Microglia are a type of glial cells that play a key role in the repair of damage to the central nervous system (CNS). In the pathological condition of Alzheimer's disease (AD), β-amyloid peptide and a variety of pro-inflammatory factors can activate microglia, resulting in the secretion of a variety of inflammatory factors and neurotoxins. This leads to neuronal damage and even apoptosis, thus triggering AD. In contrast, microglia can protect the CNS by phagocytizing Aβ to slow down AD development. In this review, the roles of microglia in AD neuroinflammation and the scope of immunotherapy for AD are summarized to provide a theoretical basis for AD prevention and treatment.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Yingying Wu
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Jichun Chen
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Aiguo Xuan
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| | - Xiao Wang
- Department of Anatomy and Neurobiology Guangzhou Medical University Guangzhou 510260 China
| |
Collapse
|
173
|
Cowan MN, Sethi I, Harris TH. Microglia in CNS infections: insights from Toxoplasma gondii and other pathogens. Trends Parasitol 2022; 38:217-229. [PMID: 35039238 PMCID: PMC8852251 DOI: 10.1016/j.pt.2021.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are poised to respond to neuropathology. Microglia play multiple roles in maintaining homeostasis and promoting inflammation in numerous disease states. The study of microglial innate immune programs has largely focused on exploring neurodegenerative disease states with the use of genetic targeting approaches. Our understanding of how microglia participate in immune responses against pathogens is just beginning to take shape. Here, we review existing animal models of CNS infection, with a focus on how microglial physiology and inflammatory processes control protozoan and viral infections of the brain. We further discuss how microglial participation in over-exuberant immune responses can drive immunopathology that is detrimental to CNS health and homeostasis.
Collapse
Affiliation(s)
- Maureen N. Cowan
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Ish Sethi
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Tajie H. Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, University of Virginia, Charlottesville, VA, United States,Correspondence: (T. H. Harris)
| |
Collapse
|
174
|
Weiss F, Labrador-Garrido A, Dzamko N, Halliday G. Immune responses in the Parkrtdinson's disease brain. Neurobiol Dis 2022; 168:105700. [DOI: 10.1016/j.nbd.2022.105700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
|
175
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
176
|
Ma Y, Yang S, He Q, Zhang D, Chang J. The Role of Immune Cells in Post-Stroke Angiogenesis and Neuronal Remodeling: The Known and the Unknown. Front Immunol 2022; 12:784098. [PMID: 34975872 PMCID: PMC8716409 DOI: 10.3389/fimmu.2021.784098] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Following a cerebral ischemic event, substantial alterations in both cellular and molecular activities occur due to ischemia-induced cerebral pathology. Mounting evidence indicates that the robust recruitment of immune cells plays a central role in the acute stage of stroke. Infiltrating peripheral immune cells and resident microglia mediate neuronal cell death and blood-brain barrier disruption by releasing inflammation-associated molecules. Nevertheless, profound immunological effects in the context of the subacute and chronic recovery phase of stroke have received little attention. Early attempts to curtail the infiltration of immune cells were effective in mitigating brain injury in experimental stroke studies but failed to exert beneficial effects in clinical trials. Neural tissue damage repair processes include angiogenesis, neurogenesis, and synaptic remodeling, etc. Post-stroke inflammatory cells can adopt divergent phenotypes that influence the aforementioned biological processes in both endothelial and neural stem cells by either alleviating acute inflammatory responses or secreting a variety of growth factors, which are substantially involved in the process of angiogenesis and neurogenesis. To better understand the multiple roles of immune cells in neural tissue repair processes post stroke, we review what is known and unknown regarding the role of immune cells in angiogenesis, neurogenesis, and neuronal remodeling. A comprehensive understanding of these inflammatory mechanisms may help identify potential targets for the development of novel immunoregulatory therapeutic strategies that ameliorate complications and improve functional rehabilitation after stroke.
Collapse
Affiliation(s)
- Yinzhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shilun Yang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qianyan He
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Dianhui Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
177
|
Zhang C, Qian X, Zheng J, Ai P, Cao X, Pan X, Chen T, Wang Y. Controlled Decompression Alleviates Brain Injury via Attenuating Oxidative Damage and Neuroinflammation in Acute Intracranial Hypertension. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1936691. [PMID: 35187159 PMCID: PMC8850036 DOI: 10.1155/2022/1936691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 01/15/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND The benefits of controlled decompression (CDC) for patients with acute intracranial hypertension especially in terms of alleviating the complications caused by rapid decompression (RDC) have been confirmed by clinical studies. This study is aimed at evaluating the therapeutic potency of CDC with ubiquitin C-terminal hydrolase-L1 (UCH-L1) and glial fibrillary acidic protein (GFAP) by investigating the potential molecular mechanism in the acute intracranial hypertension (AICH) rabbit model. METHODS Male New Zealand white rabbits were randomly subdivided into the sham-operated (SH) group, CDC group, and RDC group. Blood plasma samples and brain tissue were collected 2 days before operation (baseline) and at 3, 6, 24, and 72 hours after operation to measure the levels of UCH-L1, GFAP, oxidative stress indicators, and inflammatory cytokines by performing ELISA or Western blot. The neurological score of the rabbits and brain water content was graded 24 h after surgery. qPCR, immunofluorescence, and FJ-C staining were conducted. RESULTS CDC improved neurological function, lowered brain water content, ameliorated neuronal degeneration, attenuated oxidative damage, and inflammatory responses to a greater extent than RDC. Plasma UCH-L1 level was significantly lower in the CDC group at 3 h postoperatively than in the RDC group. CDC reduced plasma GFAP levels to various degrees at 3 h, 6 h, and 24 h postoperatively compared with RDC. Immunofluorescence confirmed that the expression of UCH-L1 and GFAP in the cortex of the CDC group was lower than that of the RDC group. CONCLUSIONS Our data collectively demonstrate that CDC could attenuate oxidative damage and inflammatory responses, downregulate UCH-L1 and GFAP levels, and contribute to an improved neuroprotective effect compared with RDC.
Collapse
Affiliation(s)
- Chonghui Zhang
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Xiao Qian
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Jie Zheng
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Pu Ai
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Xinyi Cao
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Xiaofei Pan
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Tao Chen
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Yuhai Wang
- Department of Neurosurgery, The 904th Hospital of PLA, Medical School of Anhui Medical University, Wuxi, Jiangsu 214044, China
| |
Collapse
|
178
|
Sanchez-Gonzalez R, Koupourtidou C, Lepko T, Zambusi A, Novoselc KT, Durovic T, Aschenbroich S, Schwarz V, Breunig CT, Straka H, Huttner HB, Irmler M, Beckers J, Wurst W, Zwergal A, Schauer T, Straub T, Czopka T, Trümbach D, Götz M, Stricker SH, Ninkovic J. Innate Immune Pathways Promote Oligodendrocyte Progenitor Cell Recruitment to the Injury Site in Adult Zebrafish Brain. Cells 2022; 11:520. [PMID: 35159329 PMCID: PMC8834209 DOI: 10.3390/cells11030520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 01/13/2023] Open
Abstract
The oligodendrocyte progenitors (OPCs) are at the front of the glial reaction to the traumatic brain injury. However, regulatory pathways steering the OPC reaction as well as the role of reactive OPCs remain largely unknown. Here, we compared a long-lasting, exacerbated reaction of OPCs to the adult zebrafish brain injury with a timely restricted OPC activation to identify the specific molecular mechanisms regulating OPC reactivity and their contribution to regeneration. We demonstrated that the influx of the cerebrospinal fluid into the brain parenchyma after injury simultaneously activates the toll-like receptor 2 (Tlr2) and the chemokine receptor 3 (Cxcr3) innate immunity pathways, leading to increased OPC proliferation and thereby exacerbated glial reactivity. These pathways were critical for long-lasting OPC accumulation even after the ablation of microglia and infiltrating monocytes. Importantly, interference with the Tlr1/2 and Cxcr3 pathways after injury alleviated reactive gliosis, increased new neuron recruitment, and improved tissue restoration.
Collapse
Affiliation(s)
- Rosario Sanchez-Gonzalez
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Department Biology II, University of Munich, 80539 München, Germany;
| | - Christina Koupourtidou
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Tjasa Lepko
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Alessandro Zambusi
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Klara Tereza Novoselc
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Tamara Durovic
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Sven Aschenbroich
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Veronika Schwarz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Graduate School Systemic Neurosciences, LMU, 80539 Munich, Germany
| | - Christopher T. Breunig
- Reprogramming and Regeneration, Biomedical Center (BMC), Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany; (C.T.B.); (S.H.S.)
- Epigenetic Engineering, Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany
| | - Hans Straka
- Department Biology II, University of Munich, 80539 München, Germany;
| | - Hagen B. Huttner
- Department of Neurology, Justus-Liebig-University Giessen, Klinikstrasse 33, 35392 Giessen, Germany;
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (M.I.); (J.B.)
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (M.I.); (J.B.)
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Sciences Weihenstephan, Technical University Munich, 80333 München, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (W.W.); (D.T.)
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
- Chair of Developmental Genetics c/o Helmholtz Zentrum München, School of Life Sciences Weihenstephan, Technical University Munich, 80333 München, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Munich, 80539 Munich, Germany
| | - Andreas Zwergal
- Department of Neurology, Ludwig-Maximilians University, Campus Grosshadern, 81377 Munich, Germany;
| | - Tamas Schauer
- Biomedical Center (BMC), Bioinformatic Core Facility, Faculty of Medicine, LMU Munich, 80539 München, Germany; (T.S.); (T.S.)
| | - Tobias Straub
- Biomedical Center (BMC), Bioinformatic Core Facility, Faculty of Medicine, LMU Munich, 80539 München, Germany; (T.S.); (T.S.)
| | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH8 9YL, UK;
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (W.W.); (D.T.)
| | - Magdalena Götz
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
- Biomedical Center (BMC), Division of Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany
| | - Stefan H. Stricker
- Reprogramming and Regeneration, Biomedical Center (BMC), Physiological Genomics, Faculty of Medicine, LMU Munich, 80539 München, Germany; (C.T.B.); (S.H.S.)
- Epigenetic Engineering, Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany
| | - Jovica Ninkovic
- Institute of Stem Cell Research, Helmholtz Center Munich, 85764 Oberschleißheim, Germany; (R.S.-G.); (C.K.); (T.L.); (A.Z.); (K.T.N.); (T.D.); (S.A.); (V.S.); (M.G.)
- Biomedical Center (BMC), Division of Cell Biology and Anatomy, Faculty of Medicine, LMU Munich, 80539 München, Germany
- Munich Cluster for Systems Neurology SYNERGY, LMU, 80539 Munich, Germany
| |
Collapse
|
179
|
Batra A, Bui TM, Rehring JF, Yalom LK, Muller WA, Sullivan DP, Sumagin R. Experimental Colitis Enhances Temporal Variations in CX3CR1 Cell Colonization of the Gut and Brain Following Irradiation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:295-307. [PMID: 34767810 PMCID: PMC8908021 DOI: 10.1016/j.ajpath.2021.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 02/03/2023]
Abstract
Peripheral monocyte-derived CX3C chemokine receptor 1 positive (CX3CR1+) cells play important roles in tissue homeostasis and gut repopulation. Increasing evidence also supports their role in immune repopulation of the brain parenchyma in response to systemic inflammation. Adoptive bone marrow transfer from CX3CR1 fluorescence reporter mice and high-resolution confocal microscopy was used to assess the time course of CX3CR1+ cell repopulation of steady-state and dextran sodium sulfate (DSS)-inflamed small intestine/colon and the brain over 4 weeks after irradiation. CX3CR1+ cell colonization and morphologic polarization into fully ramified cells occurred more rapidly in the small intestine than in the colon. For both organs, the crypt/mucosa was more densely populated than the serosa/muscularis layer, indicating preferential temporal and spatial occupancy. Repopulation of the brain was delayed compared with that of gut tissue, consistent with the immune privilege of this organ. However, DSS-induced colon injury accelerated the repopulation. Expression analyses confirmed increased chemokine levels and macrophage colonization within the small intestine/colon and the brain by DSS-induced injury. Early increases of transmembrane protein 119 and ionized calcium binding adaptor molecule 1 expression within the brain after colon injury suggest immune-priming effect of brain resident microglia in response to systemic inflammation. These findings identify temporal differences in immune repopulation of the gut and brain in response to inflammation and show that gut inflammation can impact immune responses within the brain.
Collapse
Affiliation(s)
- Ayush Batra
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jacob F Rehring
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lenore K Yalom
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William A Muller
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David P Sullivan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
180
|
White matter microglia heterogeneity in the CNS. Acta Neuropathol 2022; 143:125-141. [PMID: 34878590 DOI: 10.1007/s00401-021-02389-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident myeloid cells in the central nervous system (CNS) play critical roles in shaping the brain during development, responding to invading pathogens, and clearing tissue debris or aberrant protein aggregations during ageing and neurodegeneration. The original concept that like macrophages, microglia are either damaging (pro-inflammatory) or regenerative (anti-inflammatory) has been updated to a kaleidoscope view of microglia phenotypes reflecting their wide-ranging roles in maintaining homeostasis in the CNS and, their contribution to CNS diseases, as well as aiding repair. The use of new technologies including single cell/nucleus RNA sequencing has led to the identification of many novel microglia states, allowing for a better understanding of their complexity and distinguishing regional variations in the CNS. This has also revealed differences between species and diseases, and between microglia and other myeloid cells in the CNS. However, most of the data on microglia heterogeneity have been generated on cells isolated from the cortex or whole brain, whereas white matter changes and differences between white and grey matter have been relatively understudied. Considering the importance of microglia in regulating white matter health, we provide a brief update on the current knowledge of microglia heterogeneity in the white matter, how microglia are important for the development of the CNS, and how microglial ageing affects CNS white matter homeostasis. We discuss how microglia are intricately linked to the classical white matter diseases such as multiple sclerosis and genetic white matter diseases, and their putative roles in neurodegenerative diseases in which white matter is also affected. Understanding the wide variety of microglial functions in the white matter may provide the basis for microglial targeted therapies for CNS diseases.
Collapse
|
181
|
Xu L, Wang J, Ding Y, Wang L, Zhu YJ. Current Knowledge of Microglia in Traumatic Spinal Cord Injury. Front Neurol 2022; 12:796704. [PMID: 35087472 PMCID: PMC8787368 DOI: 10.3389/fneur.2021.796704] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are the resident immune cells in the central nervous system (CNS). After traumatic spinal cord injury (SCI), microglia undergo activation, proliferation, and changes in gene and protein expression and morphology, with detrimental and beneficial effects. Activated microglia cause secondary neuronal injury via the production of proinflammatory cytokines, reactive oxygen species, and proteases. However, activated microglia also promote neuronal repair through the secretion of anti-inflammatory growth factors and cytokines. Proinflammatory cytokines increase endothelial permeability, promote A1 astrocyte activation and axonal demyelination, and reduce neural stem/progenitor cells (NSPCs), leading to the exacerbation of neuronal injury. In contrast, anti-inflammatory factors facilitate angiogenesis, reduce reactive astrocytes, and promote axonal remyelination and the propagation of NSPCs, contributing to tissue repair and locomotor recovery. Due to its limited regenerative capacity, the CNS requires beneficial microglia for continuous protection against injury. Understanding and regulating microglial activation status are beneficial to reducing detrimental effects and promoting repair behaviors and to obtain more information on efficient therapies for traumatic SCI. This review discusses microglial activation and the differences between microglia and similar immune cells, microglial interactions with other cells in the spinal cord, and the progress in the development of therapies targeting microglia in SCI.
Collapse
Affiliation(s)
- Lintao Xu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyu Wang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yueming Ding
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong-Jian Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
182
|
Celorrio M, Shumilov K, Payne C, Vadivelu S, Friess SH. Acute minocycline administration reduces brain injury and improves long-term functional outcomes after delayed hypoxemia following traumatic brain injury. Acta Neuropathol Commun 2022; 10:10. [PMID: 35090569 PMCID: PMC8796448 DOI: 10.1186/s40478-022-01310-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/08/2022] [Indexed: 11/22/2022] Open
Abstract
Clinical trials of therapeutics for traumatic brain injury (TBI) demonstrating preclinical efficacy for TBI have failed to replicate these results in humans, in part due to the absence of clinically feasible therapeutic windows for administration. Minocycline, an inhibitor of microglial activation, has been shown to be neuroprotective when administered early after experimental TBI but detrimental when administered chronically to human TBI survivors. Rather than focusing on the rescue of primary injury with early administration of therapeutics which may not be clinically feasible, we hypothesized that minocycline administered at a clinically feasible time point (24 h after injury) would be neuroprotective in a model of TBI plus delayed hypoxemia. We first explored several different regimens of minocycline dosing with the initial dose 24 h after injury and 2 h prior to hypoxemia, utilizing short-term neuropathology to select the most promising candidate. We found that a short course of minocycline reduced acute microglial activation, monocyte infiltration and hippocampal neuronal loss at 1 week post injury. We then conducted a preclinical trial to assess the long-term efficacy of a short course of minocycline finding reductions in hippocampal neurodegeneration and synapse loss, preservation of white matter myelination, and improvements in fear memory performance at 6 months after injury. Timing in relation to injury and duration of minocycline treatment and its impact on neuroinflammatory response may be responsible for extensive neuroprotection observed in our studies.
Collapse
|
183
|
Soluble Endoglin Stimulates Inflammatory and Angiogenic Responses in Microglia That Are Associated with Endothelial Dysfunction. Int J Mol Sci 2022; 23:ijms23031225. [PMID: 35163148 PMCID: PMC8835690 DOI: 10.3390/ijms23031225] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1β), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFβ) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.
Collapse
|
184
|
Zhou K, Han J, Lund H, Boggavarapu NR, Lauschke VM, Goto S, Cheng H, Wang Y, Tachi A, Xie C, Zhu K, Sun Y, Osman AM, Liang D, Han W, Gemzell-Danielsson K, Betsholtz C, Zhang XM, Zhu C, Enge M, Joseph B, Harris RA, Blomgren K. An overlooked subset of Cx3cr1wt/wt microglia in the Cx3cr1CreER-Eyfp/wt mouse has a repopulation advantage over Cx3cr1CreER-Eyfp/wt microglia following microglial depletion. J Neuroinflammation 2022; 19:20. [PMID: 35062962 PMCID: PMC8783445 DOI: 10.1186/s12974-022-02381-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/28/2021] [Indexed: 01/08/2023] Open
Abstract
Background Fluorescent reporter labeling and promoter-driven Cre-recombinant technologies have facilitated cellular investigations of physiological and pathological processes, including the widespread use of the Cx3cr1CreER-Eyfp/wt mouse strain for studies of microglia. Methods Immunohistochemistry, Flow Cytometry, RNA sequencing and whole-genome sequencing were used to identify the subpopulation of microglia in Cx3cr1CreER-Eyfp/wt mouse brains. Genetically mediated microglia depletion using Cx3cr1CreER-Eyfp/wtRosa26DTA/wt mice and CSF1 receptor inhibitor PLX3397 were used to deplete microglia. Primary microglia proliferation and migration assay were used for in vitro studies. Results We unexpectedly identified a subpopulation of microglia devoid of genetic modification, exhibiting higher Cx3cr1 and CX3CR1 expression than Cx3cr1CreER-Eyfp/wtCre+Eyfp+ microglia in Cx3cr1CreER-Eyfp/wt mouse brains, thus termed Cx3cr1highCre−Eyfp− microglia. This subpopulation constituted less than 1% of all microglia under homeostatic conditions, but after Cre-driven DTA-mediated microglial depletion, Cx3cr1highCre−Eyfp− microglia escaped depletion and proliferated extensively, eventually occupying one-third of the total microglial pool. We further demonstrated that the Cx3cr1highCre−Eyfp− microglia had lost their genetic heterozygosity and become homozygous for wild-type Cx3cr1. Therefore, Cx3cr1highCre−Eyfp− microglia are Cx3cr1wt/wtCre−Eyfp−. Finally, we demonstrated that CX3CL1–CX3CR1 signaling regulates microglial repopulation both in vivo and in vitro. Conclusions Our results raise a cautionary note regarding the use of Cx3cr1CreER-Eyfp/wt mouse strains, particularly when interpreting the results of fate mapping, and microglial depletion and repopulation studies. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02381-6.
Collapse
|
185
|
Houle S, Kokiko-Cochran ON. A Levee to the Flood: Pre-injury Neuroinflammation and Immune Stress Influence Traumatic Brain Injury Outcome. Front Aging Neurosci 2022; 13:788055. [PMID: 35095471 PMCID: PMC8790486 DOI: 10.3389/fnagi.2021.788055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence demonstrates that aging influences the brain's response to traumatic brain injury (TBI), setting the stage for neurodegenerative pathology like Alzheimer's disease (AD). This topic is often dominated by discussions of post-injury aging and inflammation, which can diminish the consideration of those same factors before TBI. In fact, pre-TBI aging and inflammation may be just as critical in mediating outcomes. For example, elderly individuals suffer from the highest rates of TBI of all severities. Additionally, pre-injury immune challenges or stressors may alter pathology and outcome independent of age. The inflammatory response to TBI is malleable and influenced by previous, coincident, and subsequent immune insults. Therefore, pre-existing conditions that elicit or include an inflammatory response could substantially influence the brain's ability to respond to traumatic injury and ultimately affect chronic outcome. The purpose of this review is to detail how age-related cellular and molecular changes, as well as genetic risk variants for AD affect the neuroinflammatory response to TBI. First, we will review the sources and pathology of neuroinflammation following TBI. Then, we will highlight the significance of age-related, endogenous sources of inflammation, including changes in cytokine expression, reactive oxygen species processing, and mitochondrial function. Heightened focus is placed on the mitochondria as an integral link between inflammation and various genetic risk factors for AD. Together, this review will compile current clinical and experimental research to highlight how pre-existing inflammatory changes associated with infection and stress, aging, and genetic risk factors can alter response to TBI.
Collapse
Affiliation(s)
- Samuel Houle
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States,Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, Columbus, OH, United States
| | - Olga N. Kokiko-Cochran
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, United States,Institute for Behavioral Medicine Research, Neurological Institute, The Ohio State University, Columbus, OH, United States,*Correspondence: Olga N. Kokiko-Cochran
| |
Collapse
|
186
|
Liu Y, Fu H, Wang T. Neuroinflammation in perioperative neurocognitive disorders: From bench to the bedside. CNS Neurosci Ther 2022; 28:484-496. [PMID: 34990087 PMCID: PMC8928922 DOI: 10.1111/cns.13794] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
The perioperative neurocognitive disorders (PNDs) are one of the most common complications in elderly patients characterized by various forms of cognitive decline after anesthesia and surgery. Although the etiology for PNDs remained unclear, neuroinflammation has been characterized as one of the major causes, especially in the elderly patients. The activation of glial cells including microglia and astrocytes plays a significant role in the inflammatory responses in central nerve system (CNS). Although carefully designed, clinical studies on PNDs showed controversial results. Meanwhile, preclinical studies provided evidence from various levels, including behavior performance, protein levels, and gene expression. In this review, we summarize high‐quality studies and recent advances from both clinical and preclinical studies and provide a broad view from the onset of PNDs to its potential therapeutic targets. Future studies are needed to investigate the signaling pathways in PNDs for prevention and treatment, as well as the relationship of PNDs and future neurocognitive dysfunction.
Collapse
Affiliation(s)
- Yang Liu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Huiqun Fu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tianlong Wang
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
187
|
He X, Huang Y, Liu Y, Zhang X, Yue P, Ma X, Miao Z, Long X, Yang Y, Wan X, Lei J, Shu K, Lei T, Gan C, Zhang H. BAY61‑3606 attenuates neuroinflammation and neurofunctional damage by inhibiting microglial Mincle/Syk signaling response after traumatic brain injury. Int J Mol Med 2022; 49:5. [PMID: 34751408 PMCID: PMC8612304 DOI: 10.3892/ijmm.2021.5060] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Neuroinflammatory processes mediated by microglial activation and subsequent neuronal damage are the hallmarks of traumatic brain injury (TBI). As an inhibitor of the macrophage‑inducible C‑type lectin (Mincle)/spleen tyrosine kinase (Syk) signaling pathway, BAY61‑3606 (BAY) has previously demonstrated anti‑inflammatory effects on some pathological processes, such as acute kidney injury, by suppressing the inflammatory macrophage response. In the present study, the potential effects of BAY on microglial phenotype and neuroinflammation after TBI were investigated. BAY (3 mg/kg) was first administered into mice by intraperitoneal injection after TBI induction in vivo and microglia were also treated with BAY (2 µM) in vitro. The levels of inflammatory factors in microglia were assessed using reverse transcription‑quantitative PCR and ELISA. Cortical neuron, myelin sheath, astrocyte and cerebrovascular endothelial cell markers were detected using immunofluorescence. The levels of components of the Mincle/Syk/NF‑κB signaling pathway [Mincle, phosphorylated (p)‑Syk and NF‑κB], in addition to proteins associated with inflammation (ASC, caspase‑1, TNF‑α, IL‑1β and IL‑6), apoptosis (Bax and Bim) and tight junctions (Claudin‑5), were measured via western blotting and ELISA. Migration and chemotaxis of microglial cells were evaluated using Transwell and agarose spot assays. Neurological functions of the mice were determined in vivo using the modified neurological severity scoring system and a Morris water maze. The results of the present study revealed that the expression levels of proteins in the Mincle/Syk/NF‑κB signaling pathway (including Mincle, p‑Syk and p‑NF‑κB), inflammatory cytokines (TNF‑α, IL‑1β and IL‑6), proteins involved in inflammation (ASC and caspase‑1), apoptotic markers (Bax and Bim) and the tight junction protein Claudin‑5 were significantly altered post‑TBI. BAY treatment reversed these effects in both the cerebral cortex extract‑induced cell model and the controlled cortical impact mouse model. BAY was also revealed to suppress activation of the microglial proinflammatory phenotype and microglial migration. In addition, BAY effectively attenuated TBI‑induced neurovascular unit damage and neurological function deficits. Taken together, these findings provided evidence that BAY may inhibit the Mincle/Syk/NF‑κB signaling pathway in microglia; this in turn could attenuate microglia‑mediated neuroinflammation and improve neurological deficits following TBI.
Collapse
Affiliation(s)
- Xuejun He
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yimin Huang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xincheng Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Pengjie Yue
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaopeng Ma
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhuangzhuang Miao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaobing Long
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yiping Yang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xueyan Wan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jin Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chao Gan
- Correspondence to: Professor Huaqiu Zhang or Dr Chao Gan, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, P.R. China, E-mail: , E-mail:
| | - Huaqiu Zhang
- Correspondence to: Professor Huaqiu Zhang or Dr Chao Gan, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, P.R. China, E-mail: , E-mail:
| |
Collapse
|
188
|
Abstract
Traumatic injury of the central nervous system (CNS) is a worldwide health problem affecting millions of people. Trauma of the CNS, that is, traumatic brain injury (TBI) and spinal cord injury (SCI), lead to massive and progressive cell loss and axonal degeneration, usually with very limited regeneration. At present, there are no treatments to protect injured CNS tissue or to replace the lost tissue. Stem cells are a cell type that by definition can self-renew and give rise to multiple cell lineages. In recent years, therapies using stem and progenitor cells have shown promising effects in experimental CNS trauma, particularly in the acute-subacute stage, but also in chronic injuries. However, the therapeutic mechanisms by which transplanted cells achieve the structural and/or functional improvements are often not clear. Stem cell therapies for CNS trauma can be categorized into 2 main concepts, transplantation of exogenous neural stem cells and neural progenitor cells and recruitment of endogenous stem and progenitor cells. In this review, focusing on the advances during the last decade, we will discuss the major cell therapies, the pros and cons of these 2 concepts for TBI and SCI, and the treatment strategies we believe will be successful.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Corresponding author: Erik Sundström, Department of Neurobiology, Care Sciences and Society (NVS), BioClinicum J9:20, Karolinska University Hospital, S17164 Solna, Sweden.
| |
Collapse
|
189
|
Ponomarenko A, Tyrtyshnaia A, Ivashkevich D, Manzhulo I. Mild Traumatic Brain Injury Contributes to the Development of Delayed Neuroinflammation. Neuroimmunomodulation 2022; 29:135-142. [PMID: 34583355 DOI: 10.1159/000519011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In recent years, according to the literature, the problem of mild traumatic brain injury (mTBI) has become more and more urgent. Compared to moderate to severe craniocerebral trauma, mTBI occurs in a far greater number of people. The delayed sequelae caused by a single mTBI or multiple mTBIs are a significant public health problem. METHODS A weight-drop model was used for the formation of mTBI. A metal rod weighing 337 g with a blunt tip of 3 mm diameter was uplifted at 8 cm height and held by a lever. The trauma was created by lowering the lever and the rod and free-dropping onto the rat skull. In the cerebral cortex of experimental animals, we analyzed the level of microglial activity (Iba-1-positive system) and the expression of pro-inflammatory markers (IL1β, IL6, and CD86). Also, the expression level of the endocannabinoid system receptor (cannabinoid receptor type 1 [CB1]) was assessed in brain samples. RESULTS Experiments have shown that mTBI increases (1) the amount of microglia (iba-1) activated by the pro-inflammatory pathway (CD86); (2) the level of pro-inflammatory cytokines IL1β and IL6; and (3) CB1R activity. CONCLUSION Overall, the results of this study indicate that mTBI induces a sustained neuroinflammatory response.
Collapse
Affiliation(s)
- Arina Ponomarenko
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Anna Tyrtyshnaia
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Darya Ivashkevich
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| | - Igor Manzhulo
- Laboratory of Pharmacology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russian Federation
| |
Collapse
|
190
|
Yoshimura A, Ohyagi M, Ito M. T cells in the brain inflammation. Adv Immunol 2022; 157:29-58. [PMID: 37061287 DOI: 10.1016/bs.ai.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The immune system is deeply involved in autoimmune diseases of the central nervous system (CNS), such as multiple sclerosis, N-methyl-d-aspartate (NMDA) receptor encephalitis, and narcolepsy. Additionally, the immune system is involved in various brain diseases including cerebral infarction and neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). In particular, reports related to T cells are increasing. T cells may also play important roles in brain deterioration and dementia that occur with aging. Our understanding of the role of immune cells in the context of the brain has been greatly improved by the use of acute ischemic brain injury models. Additionally, similar neural damage and repair events are shown to occur in more chronic brain neurodegenerative brain diseases. In this review, we focus on the role of T cells, including CD4+ T cells, CD8+ T cells and regulatory T cells (Tregs) in cerebral infarction and neurodegenerative diseases.
Collapse
|
191
|
Wang YL, Wang FZ, Li R, Jiang J, Liu X, Xu J. Recent Advances in Basic Research for CSF1R-Microglial Encephalopathy. Front Aging Neurosci 2021; 13:792840. [PMID: 34955818 PMCID: PMC8695766 DOI: 10.3389/fnagi.2021.792840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
Colony-stimulating factor-1 receptor-microglial encephalopathy is a rare rapidly progressive dementia resulting from colony-stimulating factor-1 receptor (CSF1R) mutations, also named pigmentary orthochromatic leukodystrophy (POLD), hereditary diffuse leukoencephalopathy with spheroids (HDLS), adult-onset leukoencephalopathy with axonal spheroids, and pigmented glia (ALSP) and CSF1R-related leukoencephalopathy. CSF1R is primarily expressed in microglia and mutations normally directly lead to changes in microglial number and function. Many animal models have been constructed to explore pathogenic mechanisms and potential therapeutic strategies, including zebrafish, mice, and rat models which are with CSF1R monogenic mutation, biallelic or tri-allelic deletion, or CSF1R-null. Although there is no cure for patients with CSF1R-microglial encephalopathy, microglial replacement therapy has become a topical research area. This review summarizes CSF1R-related pathogenetic mutation sites and mechanisms, especially the feasibility of the microglia-original immunotherapy.
Collapse
Affiliation(s)
- Yan-Li Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fang-Ze Wang
- Department of Cardiology, Weifang People's Hospital, Weifang, China
| | - Runzhi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiangrong Liu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
192
|
Velayudhan PS, Schwab N, Hazrati LN, Wheeler AL. Temporal patterns of microglial activation in white matter following experimental mild traumatic brain injury: a systematic literature review. Acta Neuropathol Commun 2021; 9:197. [PMID: 34924026 PMCID: PMC8684664 DOI: 10.1186/s40478-021-01297-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are a prevalent form of injury that can result in persistent neurological impairments. Microglial activation has become increasingly recognized as a key process regulating the pathology of white matter in a wide range of brain injury and disease contexts. As white matter damage is known to be a major contributor to the impairments that follow mTBI, microglia have rightfully become a common target of investigation for the development of mTBI therapies and biomarkers. Recent work has demonstrated that the efficacy of microglial manipulation as a therapeutic intervention following injury or disease is highly time-sensitive, emphasizing the importance of advancing our understanding of the dynamics of post-mTBI microglial activation from onset to resolution. Current reporting of microglial activation in experimental studies of mTBI is non-standardized, which has limited our ability to identify concrete patterns of post-mTBI microglial activation over time. In this review, we examine preclinical studies of mTBI that report on microglial activation in white matter regions to summarize our current understanding of these patterns. Specifically, we summarize timecourses of post-mTBI microglial activation in white matter regions of the brain, identify factors that influence this activation, examine the temporal relationship between microglial activation and other post-mTBI assessments, and compare the relative sensitivities of various methods for detecting microglial activation. While the lack of replicated experimental conditions has limited the extent of conclusions that can confidently be drawn, we find that microglia are activated over a wide range of timecourses following mTBI and that microglial activation is a long-lasting outcome of mTBI that may resolve after most typical post-mTBI assessments, with the exception of those measuring oligodendrocyte lineage cell integrity. We identify several understudied parameters of post-mTBI microglial activation in white matter, such as the inclusion of female subjects. This review summarizes our current understanding of the progression of microglial activation in white matter structures following experimental mTBI and offers suggestions for important future research directions.
Collapse
Affiliation(s)
- Prashanth S Velayudhan
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Nicole Schwab
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Lili-Naz Hazrati
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anne L Wheeler
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
193
|
Zhang L, Xu H, Ding N, Li X, Chen X, Chen Z. Beneficial Effects on Brain Micro-Environment by Caloric Restriction in Alleviating Neurodegenerative Diseases and Brain Aging. Front Physiol 2021; 12:715443. [PMID: 34899367 PMCID: PMC8660583 DOI: 10.3389/fphys.2021.715443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Aging and neurodegenerative diseases are frequently associated with the disruption of the extracellular microenvironment, which includes mesenchyme and body fluid components. Caloric restriction (CR) has been recognized as a lifestyle intervention that can improve long-term health. In addition to preventing metabolic disorders, CR has been shown to improve brain health owing to its enhancing effect on cognitive functions or retarding effect on the progression of neurodegenerative diseases. This article summarizes current findings regarding the neuroprotective effects of CR, which include the modulation of metabolism, autophagy, oxidative stress, and neuroinflammation. This review may offer future perspectives for brain aging interventions.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, China.,Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ning Ding
- Key Laboratory of Central CNS Regeneration (Ministry of Education), Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Medical College, Kunming University of Science and Technology, Kunming, China
| | - Xue Li
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhuangfei Chen
- Medical College, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
194
|
Hu Y, Tao W. Microenvironmental Variations After Blood-Brain Barrier Breakdown in Traumatic Brain Injury. Front Mol Neurosci 2021; 14:750810. [PMID: 34899180 PMCID: PMC8662751 DOI: 10.3389/fnmol.2021.750810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is linked to several pathologies. The blood-brain barrier (BBB) breakdown is considered to be one of the initial changes. Further, the microenvironmental alteration following TBI-induced BBB breakdown can be multi-scaled, constant, and dramatic. The microenvironmental variations after disruption of BBB includes several pathological changes, such as cerebral blood flow (CBF) alteration, brain edema, cerebral metabolism imbalances, and accumulation of inflammatory molecules. The modulation of the microenvironment presents attractive targets for TBI recovery, such as reducing toxic substances, inhibiting inflammation, and promoting neurogenesis. Herein, we briefly review the pathological alterations of the microenvironmental changes following BBB breakdown and outline potential interventions for TBI recovery based on microenvironmental modulation.
Collapse
Affiliation(s)
- Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
195
|
Beneficial and detrimental functions of microglia during viral encephalitis. Trends Neurosci 2021; 45:158-170. [PMID: 34906391 DOI: 10.1016/j.tins.2021.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/28/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022]
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with multiple functions in health and disease. Their response during encephalitis depends on whether inflammation is triggered in a sterile or infectious manner, and in the latter case on the type of the infecting pathogen. Even though recent technological innovations advanced the understanding of the broad spectrum of microglia responses during viral encephalitis (VE), it is not entirely clear which microglia gene expression profiles are associated with antiviral and detrimental activities. Here, we review novel approaches to study microglia and the latest concepts of their function in VE. Improved understanding of microglial functions will be essential for the development of new therapeutic interventions for VE.
Collapse
|
196
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|
197
|
Chu E, Mychasiuk R, Hibbs ML, Semple BD. Dysregulated phosphoinositide 3-kinase signaling in microglia: shaping chronic neuroinflammation. J Neuroinflammation 2021; 18:276. [PMID: 34838047 PMCID: PMC8627624 DOI: 10.1186/s12974-021-02325-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia are integral mediators of innate immunity within the mammalian central nervous system. Typical microglial responses are transient, intending to restore homeostasis by orchestrating the removal of pathogens and debris and the regeneration of damaged neurons. However, prolonged and persistent microglial activation can drive chronic neuroinflammation and is associated with neurodegenerative disease. Recent evidence has revealed that abnormalities in microglial signaling pathways involving phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT) may contribute to altered microglial activity and exacerbated neuroimmune responses. In this scoping review, the known and suspected roles of PI3K-AKT signaling in microglia, both during health and pathological states, will be examined, and the key microglial receptors that induce PI3K-AKT signaling in microglia will be described. Since aberrant signaling is correlated with neurodegenerative disease onset, the relationship between maladapted PI3K-AKT signaling and the development of neurodegenerative disease will also be explored. Finally, studies in which microglial PI3K-AKT signaling has been modulated will be highlighted, as this may prove to be a promising therapeutic approach for the future treatment of a range of neuroinflammatory conditions.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Immunology and Pathology, Central Clinical School, Monash University, Level 6, 89 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Prahran, VIC, 3181, Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Level 6, 89 Commercial Road, Melbourne, VIC, 3004, Australia.
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Prahran, VIC, 3181, Australia.
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, 3050, Australia.
| |
Collapse
|
198
|
Tsai MS, Wang LC, Tsai HY, Lin YJ, Wu HL, Tzeng SF, Hsu SM, Chen SH. Microglia Reduce Herpes Simplex Virus 1 Lethality of Mice with Decreased T Cell and Interferon Responses in Brains. Int J Mol Sci 2021; 22:ijms222212457. [PMID: 34830340 PMCID: PMC8624831 DOI: 10.3390/ijms222212457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 12/11/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) infects the majority of the human population and can induce encephalitis, which is the most common cause of sporadic, fatal encephalitis. An increase of microglia is detected in the brains of encephalitis patients. The issues regarding whether and how microglia protect the host and neurons from HSV-1 infection remain elusive. Using a murine infection model, we showed that HSV-1 infection on corneas increased the number of microglia to outnumber those of infiltrating leukocytes (macrophages, neutrophils, and T cells) and enhanced microglia activation in brains. HSV-1 antigens were detected in brain neurons, which were surrounded by microglia. Microglia depletion increased HSV-1 lethality of mice with elevated brain levels of viral loads, infected neurons, neuron loss, CD4 T cells, CD8 T cells, neutrophils, interferon (IFN)-β, and IFN-γ. In vitro studies demonstrated that microglia from infected mice reduced virus infectivity. Moreover, microglia induced IFN-β and the signaling pathway of signal transducer and activator of transcription (STAT) 1 to inhibit viral replication and damage of neurons. Our study reveals how microglia protect the host and neurons from HSV-1 infection.
Collapse
Affiliation(s)
- Meng-Shan Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
| | - Li-Chiu Wang
- School of Medicine, I-Shou University, Kaohsiung 824, Taiwan;
| | - Hsien-Yang Tsai
- Department of Ophthalmology, Tzu Chi Hospital, Taichung 427, Taiwan;
| | - Yu-Jheng Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Biological Science and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan;
| | - Sheng-Min Hsu
- Department of Ophthalmology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (S.-M.H.); (S.-H.C.)
| | - Shun-Hua Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; (M.-S.T.); (H.-L.W.)
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Correspondence: (S.-M.H.); (S.-H.C.)
| |
Collapse
|
199
|
Lemon N, Canepa E, Ilies MA, Fossati S. Carbonic Anhydrases as Potential Targets Against Neurovascular Unit Dysfunction in Alzheimer’s Disease and Stroke. Front Aging Neurosci 2021; 13:772278. [PMID: 34867298 PMCID: PMC8635164 DOI: 10.3389/fnagi.2021.772278] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/20/2021] [Indexed: 12/23/2022] Open
Abstract
The Neurovascular Unit (NVU) is an important multicellular structure of the central nervous system (CNS), which participates in the regulation of cerebral blood flow (CBF), delivery of oxygen and nutrients, immunological surveillance, clearance, barrier functions, and CNS homeostasis. Stroke and Alzheimer Disease (AD) are two pathologies with extensive NVU dysfunction. The cell types of the NVU change in both structure and function following an ischemic insult and during the development of AD pathology. Stroke and AD share common risk factors such as cardiovascular disease, and also share similarities at a molecular level. In both diseases, disruption of metabolic support, mitochondrial dysfunction, increase in oxidative stress, release of inflammatory signaling molecules, and blood brain barrier disruption result in NVU dysfunction, leading to cell death and neurodegeneration. Improved therapeutic strategies for both AD and stroke are needed. Carbonic anhydrases (CAs) are well-known targets for other diseases and are being recently investigated for their function in the development of cerebrovascular pathology. CAs catalyze the hydration of CO2 to produce bicarbonate and a proton. This reaction is important for pH homeostasis, overturn of cerebrospinal fluid, regulation of CBF, and other physiological functions. Humans express 15 CA isoforms with different distribution patterns. Recent studies provide evidence that CA inhibition is protective to NVU cells in vitro and in vivo, in models of stroke and AD pathology. CA inhibitors are FDA-approved for treatment of glaucoma, high-altitude sickness, and other indications. Most FDA-approved CA inhibitors are pan-CA inhibitors; however, specific CA isoforms are likely to modulate the NVU function. This review will summarize the literature regarding the use of pan-CA and specific CA inhibitors along with genetic manipulation of specific CA isoforms in stroke and AD models, to bring light into the functions of CAs in the NVU. Although pan-CA inhibitors are protective and safe, we hypothesize that targeting specific CA isoforms will increase the efficacy of CA inhibition and reduce side effects. More studies to further determine specific CA isoforms functions and changes in disease states are essential to the development of novel therapies for cerebrovascular pathology, occurring in both stroke and AD.
Collapse
Affiliation(s)
- Nicole Lemon
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Elisa Canepa
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Marc A. Ilies
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, United States
| | - Silvia Fossati
- Alzheimer’s Center at Temple (ACT), Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- *Correspondence: Silvia Fossati,
| |
Collapse
|
200
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|