51
|
Abstract
Stroke is a sudden and rapidly progressing ischemic or hemorrhagic cerebrovascular disease. When stroke damages the brain, the immune system becomes hyperactive, leading to systemic inflammatory response and immunomodulatory disorders, which could significantly impact brain damage, recovery, and prognosis of stroke. Emerging researches suggest that ischemic stroke-induced spleen contraction could activate a peripheral immune response, which may further aggravate brain injury. This review focuses on hemorrhagic strokes including intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH) and discusses the central nervous system-peripheral immune interactions after hemorrhagic stroke induction. First, inflammatory progression after ICH and SAH is investigated. As a part of this review, we summarize the various kinds of inflammatory cell infiltration to aggravate brain injury after blood-brain barrier interruption induced by hemorrhagic stroke. Then, we explore hemorrhagic stroke-induced systemic inflammatory response syndrome (SIRS) and discuss the interactions of CNS and peripheral inflammatory response. In addition, potential targets related to inflammatory response for ICH and SAH are discussed in this review, which may lead to novel therapeutic strategies for hemorrhagic stroke.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Stroke Research, Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Stroke Research, Soochow University, Suzhou, China
| |
Collapse
|
52
|
Zhang W, Wu Q, Hao S, Chen S. The hallmark and crosstalk of immune cells after intracerebral hemorrhage: Immunotherapy perspectives. Front Neurosci 2023; 16:1117999. [PMID: 36711145 PMCID: PMC9877537 DOI: 10.3389/fnins.2022.1117999] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most dangerous types of strokes with a high morbidity and mortality rate. Currently, the treatment of ICH is not well developed, mainly because its mechanisms are still unclear. Inflammation is one of the main types of secondary injury after ICH and catalyzes the adverse consequences of ICH. A large number of immune cells are involved in neuroinflammation, such as microglia, astrocytes, oligodendrocytes, lymphocytes, macrophages, and neutrophils. Nevertheless, the characteristics and crosstalk of immune cells have not been fully elucidated. In this review, we endeavor to delve into the respective characteristics of immune cells and their interactions in neuroimmune inflammation, and further elucidate favorable immunotherapeutic approaches regarding ICH, and finally present an outlook.
Collapse
Affiliation(s)
- Wenqing Zhang
- School of Medicine, Chongqing University, Chongqing, China,Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,*Correspondence: Shilei Hao,
| | - Shengli Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China,Shengli Chen,
| |
Collapse
|
53
|
Zhu M, Li S, Cao X, Rashid K, Liu T. The STAT family: Key transcription factors mediating crosstalk between cancer stem cells and tumor immune microenvironment. Semin Cancer Biol 2023; 88:18-31. [PMID: 36410636 DOI: 10.1016/j.semcancer.2022.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Signal transducer and activator of transcription (STAT) proteins compose a family of transcription factors critical for cancer stem cells (CSCs), and they are involved in maintaining stemness properties, enhancing cell proliferation, and promoting metastasis. Recent studies suggest that STAT proteins engage in reciprocal communication between CSCs and infiltrate immune cell populations in the tumor microenvironment (TME). Emerging evidence has substantiated the influence of immune cells, including macrophages, myeloid-derived suppressor cells, and T cells, on CSC survival through the regulation of STAT signaling. Conversely, dysregulation of STATs in CSCs or immune cells contributes to the establishment of an immunosuppressive TME. Thus, STAT proteins are promising therapeutic targets for cancer treatment, especially when used in combination with immunotherapy. From this perspective, we discuss the complex roles of STATs in CSCs and highlight their functions in the crosstalk between CSCs and the immune microenvironment. Finally, cutting-edge clinical trial progress with STAT signaling inhibitors is summarized.
Collapse
Affiliation(s)
- Mengxuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China; Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Suyao Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China; Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Khalid Rashid
- Department of Cancer Biology, Faculty of Medicine, University of Cincinnati, OH, USA.
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China; Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
54
|
Batts AJ, Ji R, Noel RL, Kline-Schoder AR, Bae S, Kwon N, Konofagou EE. Using a novel rapid alternating steering angles pulse sequence to evaluate the impact of theranostic ultrasound-mediated ultra-short pulse length on blood-brain barrier opening volume and closure, cavitation mapping, drug delivery feasibility, and safety. Theranostics 2023; 13:1180-1197. [PMID: 36793858 PMCID: PMC9925313 DOI: 10.7150/thno.76199] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/08/2022] [Indexed: 02/16/2023] Open
Abstract
Background: Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is a noninvasive, safe and reversible technique for targeted drug delivery to the brain. Most preclinical systems developed to perform and monitor BBB opening are comprised of a separate geometrically focused transducer and passive cavitation detector (PCD) or imaging array. This study builds upon previous work from our group developing a single imaging phased array configuration for simultaneous BBB opening and monitoring called theranostic ultrasound (ThUS), leveraging ultra-short pulse lengths (USPLs) and a novel rapid alternating steering angles (RASTA) pulse sequence design for simultaneous bilateral sonications with target-specific USPL. The RASTA sequence was further employed to evaluate the impact of USPL on BBB opening volume, power cavitation imaging (PCI) pixel intensity, BBB closing timeline, drug delivery efficiency, and safety. Methods: A P4-1 phased array transducer driven by a Verasonics Vantage ultrasound system was operated using a custom script to run the RASTA sequence which consisted of interleaved steered, focused transmits and passive imaging. Contrast-enhanced magnetic resonance imaging (MRI) confirmed initial opening volume and closure of the BBB by longitudinal imaging through 72 hours post-BBB opening. For drug delivery experiments, mice were systemically administered a 70 kDa fluorescent dextran or adeno-associated virus serotype 9 (AAV9) for fluorescence microscopy or enzyme-linked immunosorbent assay (ELISA) to evaluate ThUS-mediated molecular therapeutic delivery. Additional brain sections were also H&E-stained to evaluate histological damage, and IBA1- and GFAP-stained to elucidate the effects of ThUS-mediated BBB opening on stimulation of key cell types involved in the neuro-immune response, microglia and astrocytes. Results: The ThUS RASTA sequence induced distinct BBB openings simultaneously in the same mouse where volume, PCI pixel intensity, level of dextran delivery, and AAV reporter transgene expression were correlated with brain hemisphere-specific USPL, consistent with statistically significant differences between 1.5, 5, and 10-cycle USPL groups. BBB closure after ThUS required 2-48 hours depending on USPL. The potential for acute damage and neuro-immune activation increased with USPL, but such observable damage was nearly reversed 96 hours post-ThUS. Conclusion: ThUS is a versatile single-array technique which exhibits the potential for investigating a variety of non-invasive therapeutic delivery applications in the brain.
Collapse
Affiliation(s)
- Alec J Batts
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Rebecca L Noel
- Department of Biomedical Engineering, Columbia University, New York, USA
| | | | - Sua Bae
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, USA.,Department of Radiology, Columbia University, New York, USA
| |
Collapse
|
55
|
Wan Y, Holste KG, Hua Y, Keep RF, Xi G. Brain edema formation and therapy after intracerebral hemorrhage. Neurobiol Dis 2023; 176:105948. [PMID: 36481437 PMCID: PMC10013956 DOI: 10.1016/j.nbd.2022.105948] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/28/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) accounts for about 10% of all strokes in the United States of America causing a high degree of disability and mortality. There is initial (primary) brain injury due to the mechanical disruption caused by the hematoma. There is then secondary injury, triggered by the initial injury but also the release of various clot-derived factors (e.g., thrombin and hemoglobin). ICH alters brain fluid homeostasis. Apart from the initial hematoma mass, ICH causes blood-brain barrier disruption and parenchymal cell swelling, which result in brain edema and intracranial hypertension affecting patient prognosis. Reducing brain edema is a critical part of post-ICH care. However, there are limited effective treatment methods for reducing perihematomal cerebral edema and intracranial pressure in ICH. This review discusses the mechanisms underlying perihematomal brain edema formation, the effects of sex and age, as well as how edema is resolved. It examines progress in pharmacotherapy, particularly focusing on drugs which have been or are currently being investigated in clinical trials.
Collapse
Affiliation(s)
- Yingfeng Wan
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
56
|
Li Y, Tian C, Wei Y, Liu H, An N, Song K, Sun Y, Gao Y, Gao Y. Exploring the pharmacological mechanism of Naoxueshu oral liquid in the treatment of intracerebral hemorrhage through weighted gene co-expression network analysis, network pharmacological and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154530. [PMID: 36356328 DOI: 10.1016/j.phymed.2022.154530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a life-threatening stroke subtype with high rates of disability and mortality. Naoxueshu oral liquid is a proprietary Chinese medicine that absorbs hematoma and exhibits neuroprotective effects in patients with ICH. However, the underlying mechanisms remain obscure. PURPOSE Exploring and elucidating the pharmacological mechanism of Naoxueshu oral liquid in the treatment of ICH. STUDY DESIGN AND METHODS The Gene Expression Omnibus (GEO) database was used to download the gene expression data on ICH. ICH-related hub modules were obtained by weighted gene co-expression network analysis (WGCNA) of differentially co-expressed genes (DEGs). The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the obtained key modules to identify the ICH-related signaling pathways. Network pharmacology technology was applied to forecast the targets of Naoxueshu oral liquid and to establish a protein-protein interaction (PPI) network of overlapping targets between Naoxueshu oral liquid and ICH. Functional annotation and enrichment pathway analyses of the intersectional targets were performed using the omicsbean database. Finally, we verified the therapeutic role and mechanism of Naoxueshu oral liquid in ICH through molecular docking and experiments. RESULTS Through the WGCNA analysis, combined with network pharmacology, it was found that immune inflammation was closely related to the early pathological mechanism of ICH. Naoxueshu oral liquid suppressed the inflammatory response; hence, it could be a potential drug for ICH treatment. Molecular docking further confirmed that the effective components of Naoxueshu oral liquid docked well with CD163. Finally, the experimental results showed that Naoxueshu oral liquid treatment in the ICH rat model attenuated neurological deficits and neuronal injury, decreased hematoma volume, and promoted hematoma absorption. In addition, Naoxueshu oral liquid treatment also significantly increased the levels of Arg-1, CD163, Nrf2, and HO-1 around hematoma after ICH. CONCLUSION This study demonstrated that Naoxueshu oral liquid attenuated neurological deficits and accelerated hematoma absorption, possibly by suppressing inflammatory responses, which might be related to the regulation of Nrf2/CD163/HO-1 that interfered with the activation of M2 microglia, thus accelerating the clearance and decomposition of hemoglobin in the hematoma.
Collapse
Affiliation(s)
- Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Tian
- Beijing University of Chinese Medicine, Beijing, 100029, China; China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yufei Wei
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Guangxi, 530000, China
| | - Haoqi Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ke Song
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Ying Gao
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
57
|
Liu C, Li Z, Xi H. Bioinformatics analysis and in vivo validation of ferroptosis-related genes in ischemic stroke. Front Pharmacol 2022; 13:940260. [PMID: 36506580 PMCID: PMC9729703 DOI: 10.3389/fphar.2022.940260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Ischemic stroke (IS) is a neurological condition associated with high mortality and disability rates. Although the molecular mechanisms underlying IS remain unclear, ferroptosis was shown to play an important role in its pathogenesis. Hence, we applied bioinformatics analysis to identify ferroptosis-related therapeutic targets in IS. IS-related microarray data from the GSE61616 dataset were downloaded from the Gene Expression Omnibus (GEO) database and intersected with the FerrDb database. In total, 33 differentially expressed genes (DEGs) were obtained and subjected to functional enrichment and protein-protein interaction (PPI) network analyses. Four candidate genes enriched in the HIF-1 signaling pathway (HMOX1, STAT3, CYBB, and TLR4) were selected based on the hierarchical clustering of the PPI dataset. We also downloaded the IR-related GSE35338 dataset and GSE58294 dataset from the GEO database to verify the expression levels of these four genes. ROC monofactor analysis demonstrated a good performance of HMOX1, STAT3, CYBB, and TLR4 in the diagnosis of ischemic stroke. Transcriptional levels of the above four genes, and translational level of GPX4, the central regulator of ferroptosis, were verified in a mouse model of middle cerebral artery occlusion (MCAO)-induced IS by qRT-PCR and western blotting. Considering the regulation of the HIF-1 signaling pathway, dexmedetomidine was applied to the MCAO mice. We found that expression of these four genes and GPX4 in MCAO mice were significantly reduced, while dexmedetomidine reversed these changes. In addition, dexmedetomidine significantly reduced MCAO-induced cell death, improved neurobehavioral deficits, and reduced the serum and brain levels of inflammatory factors (TNF-α and IL-6) and oxidative stress mediators (MDA and GSSG). Further, we constructed an mRNA-miRNA-lncRNA network based on the four candidate genes and predicted possible transcription factors. In conclusion, we identified four ferroptosis-related candidate genes in IS and proposed, for the first time, a possible mechanism for dexmedetomidine-mediated inhibition of ferroptosis during IS. These findings may help design novel therapeutic strategies for the treatment of IS.
Collapse
Affiliation(s)
- Chang Liu
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China,The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China,Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhixi Li
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China,The Key Laboratory of Myocardial Ischemia Organization, Chinese Ministry of Education, Harbin, China,Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongjie Xi
- Heilongjiang Province Key Laboratory of Research on Anesthesiology and Critical Care Medicine, Harbin, China,Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,*Correspondence: Hongjie Xi,
| |
Collapse
|
58
|
Paiva WS, Zippo E, Miranda C, Brasil S, Godoy DA, De Andrade AF, Neville I, Patriota GC, Domingues R, Teixeira MJ. Animal models for the study of intracranial hematomas (Review). Exp Ther Med 2022; 25:20. [PMID: 36561628 PMCID: PMC9748783 DOI: 10.3892/etm.2022.11719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Intracranial hematomas (ICH) are a frequent condition in neurosurgical and neurological practices, with several mechanisms of primary and secondary injury. Experimental research has been fundamental for the understanding of the pathophysiology implicated with ICH and the development of therapeutic interventions. To date, a variety of different animal approaches have been described that consider, for example, the ICH evolutive phase, molecular implications and hemodynamic changes. Therefore, choosing a test protocol should consider the scope of each particular study. The present review summarized investigational protocols in experimental research on the subject of ICH. With this subject, injection of autologous blood or bacterial collagenase, inflation of intracranial balloon and avulsion of cerebral vessels were the models identified. Rodents (mice) and swine were the most frequent species used. These different models allowed improvements on the understanding of intracranial hypertension establishment, neuroinflammation, immunology, brain hemodynamics and served to the development of therapeutic strategies.
Collapse
Affiliation(s)
- Wellingson Silva Paiva
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil,Medical Research Laboratory 62, Department of Neurology, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil
| | - Emanuele Zippo
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil
| | - Carolina Miranda
- Neurology Center, Samaritan Hospital, 01232010 São Paulo, Brazil
| | - Sérgio Brasil
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil,Medical Research Laboratory 62, Department of Neurology, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil,Correspondence to: Dr Sérgio Brasil, Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 255 Enéas Aguiar Street, 05403 São Paulo, Brazil
| | - Daniel Augustin Godoy
- Department of Intensive Care, Neurointensive Care Unit, Pasteur Hospital, 4700 Catamarca, Argentina
| | - Almir Ferreira De Andrade
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil,Medical Research Laboratory 62, Department of Neurology, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil
| | - Iuri Neville
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil
| | | | - Renan Domingues
- Neurology Center, Samaritan Hospital, 01232010 São Paulo, Brazil
| | - Manoel Jacobsen Teixeira
- Department of Neurology, Division of Neurosurgery, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil,Medical Research Laboratory 62, Department of Neurology, School of Medicine, University of São Paulo, 05403000 São Paulo, Brazil
| |
Collapse
|
59
|
Luo J, Chen Y, Tang G, Li Z, Yang X, Shang X, Huang T, Huang G, Wang L, Han Y, Zhou Y, Wang C, Wu B, Guo Q, Gong B, Li M, Wang R, Yang J, Cui W, Zhong J, Zhong LL, Guo J. Gut microbiota composition reflects disease progression, severity and outcome, and dysfunctional immune responses in patients with hypertensive intracerebral hemorrhage. Front Immunol 2022; 13:869846. [PMID: 36439158 PMCID: PMC9699794 DOI: 10.3389/fimmu.2022.869846] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/08/2022] [Indexed: 07/28/2023] Open
Abstract
OBJECTIVE In this study, we aimed to explore the alterations in gut microbiota composition and cytokine responses related to disease progression, severity, and outcomes in patients with hypertensive intracerebral hemorrhage (ICH). METHODS Fecal microbiota communities of 64 patients with ICH, 46 coronary heart disease controls, and 23 healthy controls were measured by sequencing the V3-V4 region of the 16S ribosomal RNA (16S rRNA) gene. Serum concentrations of a broad spectrum of cytokines were examined by liquid chips and ELISA. Relationships between clinical phenotypes, microbiotas, and cytokine responses were analyzed in the group with ICH and stroke-associated pneumonia (SAP), the major complication of ICH. RESULTS In comparison with the control groups, the gut microbiota of the patients with ICH had increased microbial richness and diversity, an expanded spectrum of facultative anaerobes and opportunistic pathogens, and depletion of anaerobes. Enterococcus enrichment and Prevotella depletion were more significant in the ICH group and were associated with the severity and functional outcome of ICH. Furthermore, Enterococcus enrichment and Prevotella depletion were also noted in the SAP group in contrast to the non-SAP group. Enterococci were also promising factors in the prognosis of ICH. The onset of ICH induced massive, rapid activation of the peripheral immune system. There were 12 cytokines (Eotaxin, GM-CSF, IL-8, IL-9, IL-10, IL-12p70, IL-15, IL-23, IL-1RA, IP-10, RANTES, and TNF-α) changed significantly with prolongation of ICH, and the Th2 responses correlated with the 90-day outcomes. Cytokines TNF-α, IP-10, IL-1RA, IL-8, IL-18, and MIP-1β in SAP group significantly differed from non-SAP group. Among these cytokines, only IP-10 levels decreased in the SAP group. Enterococcus was positively associated with IL-1RA and negatively associated with IP-10, while Prevotella was inversely associated in both the ICH and SAP groups. CONCLUSION This study revealed that gut dysbiosis with enriched Enterococcus and depleted Prevotella increased the risk of ICH and subsequently SAP. The altered gut microbiota composition and serum cytokine profiles are potential biomarkers that reflect the inciting physiologic insult/stress involved with ICH.
Collapse
Affiliation(s)
- Jielian Luo
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yang Chen
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guanghai Tang
- Department of Neurology, Shenyang Second Hospital of Traditional Chinese Medicine, Shenyang, China
| | - Zhuo Li
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Genetic Testing Lab, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobo Yang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Xiaoxiao Shang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tao Huang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gan Huang
- Department of Neurology, Yangjiang Hospital of Traditional Chinese Medicine, Yangjiang, China
| | - Lixin Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun Han
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuexiang Zhou
- Department of Community Healthcare Service, Shenzhen FuYong People’s Hospital, Shenzhen, China
| | - Chuyang Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Wu
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Genetic Testing Lab, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qihua Guo
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Baoying Gong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Mengzhen Li
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ruihua Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Fourth Affiliated Hospital of Guangzhou Medical University Research Team of Traditional Chinese Medicine for the Prevention and Treatment of Cerebral Hemorrhage, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiecong Yang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Wanzhen Cui
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jianbin Zhong
- Department of Neurology, The Fourth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linda Ld Zhong
- Hong Kong Chinese Medicine Clinical Study Centre, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jianwen Guo
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
60
|
Yang G, Fan X, Mazhar M, Guo W, Zou Y, Dechsupa N, Wang L. Neuroinflammation of microglia polarization in intracerebral hemorrhage and its potential targets for intervention. Front Mol Neurosci 2022; 15:1013706. [PMID: 36304999 PMCID: PMC9592761 DOI: 10.3389/fnmol.2022.1013706] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a key role in neurological diseases, including intracerebral hemorrhage (ICH). Microglia are activated to acquire either pro-inflammatory or anti-inflammatory phenotypes. After the onset of ICH, pro-inflammatory mediators produced by microglia at the early stages serve as a crucial character in neuroinflammation. Conversely, switching the microglial shift to an anti-inflammatory phenotype could alleviate inflammatory response and incite recovery. This review will elucidate the dynamic profiles of microglia phenotypes and their available shift following ICH. This study can facilitate an understanding of the self-regulatory functions of the immune system involving the shift of microglia phenotypes in ICH. Moreover, suggestions for future preclinical and clinical research and potential intervention strategies are discussed.
Collapse
Affiliation(s)
- Guoqiang Yang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- First Department of Medicine, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Wubin Guo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yuanxia Zou
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- *Correspondence: Li Wang Nathupakorn Dechsupa
| | - Li Wang
- Research Center for Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
- *Correspondence: Li Wang Nathupakorn Dechsupa
| |
Collapse
|
61
|
Xia F, Keep RF, Ye F, Holste KG, Wan S, Xi G, Hua Y. The Fate of Erythrocytes after Cerebral Hemorrhage. Transl Stroke Res 2022; 13:655-664. [PMID: 35066815 PMCID: PMC9782724 DOI: 10.1007/s12975-021-00980-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 02/05/2023]
Abstract
After a cerebral hemorrhage (intracerebral, subarachnoid, and intraventricular), extravasated blood contributes to both initial brain injury, via physical disruption and mass effect, and secondary injury, through the release of potentially neurotoxic and pro-inflammatory factors such as hemoglobin, iron, and peroxiredoxin-2. Erythrocytes are a major blood component and are a source of such damaging factors. Erythrolysis after cerebral hemorrhage releases potential neurotoxins, contributing to brain injury and edema. Alternatively, erythrocyte phagocytosis via microglia or macrophages may limit the spill of neurotoxins therefore limiting subsequent brain injury. The aim of this review is to discuss the process of phagocytosis of erythrocytes by microglia or macrophages after cerebral hemorrhage, the effect of erythrolysis on brain injury, novel mechanisms of erythrocyte and phagocyte egress from the brain, and exciting new targets in this pathway to attenuate brain injury. Understanding the fate of erythrocytes after cerebral hemorrhage may uncover additional potential interventions for clinical translational research.
Collapse
Affiliation(s)
- Fan Xia
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Katherine G Holste
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Shu Wan
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
62
|
Sun T, Zeng H, Fan L, Fei J, Chen G. Semaphorin 6D regulate corralling, hematoma compaction and white matter injury in mice after intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2022; 31:106803. [PMID: 36174325 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES The Semaphorin 6D (SEMA6D) shows important roles in cell guidance and lipid metabolism, but the effects and mechanisms of SEMA6D on tissue repair, white matter injury and the recovery of neurological function after intracerebral hemorrhage have not been well studied. MATERIALS AND METHODS In this study, the autologous whole blood injection model of intracerebral hemorrhage was established in C57 male mice. SEMA6D knockout CRISPR utilized in the study. Assessments included neurological score evaluation and immunofluorescence. RESULTS SEMA6D increased and peaked at 7d after intracerebral hemorrhage, and mainly located in neurons, microglia and astrocytes. SEMA6D knockout CRISPR aggravated neurological function and showed signs of poorer corralling and hematoma resolution, with more compartments of well-established physical barrier and more extensive GFAP positive astrocytic border. Furthermore, SEMA6D can prevent the decrease of NF-H in the peri-hematoma region, while SEMA6D knockout aggravated WMI. CONCLUSIONS Our study suggested that SEMA6D could influence the recovery of neurological function by regulating the corralling, hematoma compaction and WMI in mice after intracerebral hemorrhage.
Collapse
Affiliation(s)
- Ting Sun
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Hanhai Zeng
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Linfeng Fan
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| | - Jing Fei
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China.
| | - Gao Chen
- Department of Neurological Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, Zhejiang Province 310009, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China..
| |
Collapse
|
63
|
Zheng Y, Li R, Fan X. Targeting Oxidative Stress in Intracerebral Hemorrhage: Prospects of the Natural Products Approach. Antioxidants (Basel) 2022; 11:1811. [PMID: 36139885 PMCID: PMC9495708 DOI: 10.3390/antiox11091811] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH), the second most common subtype of stroke, remains a significant cause of morbidity and mortality worldwide. The pathological mechanism of ICH is very complex, and it has been demonstrated that oxidative stress (OS) plays an important role in the pathogenesis of ICH. Previous studies have shown that OS is a therapeutic target after ICH, and antioxidants have also achieved some benefits in the treatment of ICH. This review aimed to explore the promise of natural products therapy to target OS in ICH. We searched PubMed using the keywords "oxidative stress in intracerebral hemorrhage" and "natural products in intracerebral hemorrhage". Numerous animal and cell studies on ICH have demonstrated the potent antioxidant properties of natural products, including polyphenols and phenolic compounds, terpenoids, alkaloids, etc. In summary, natural products such as antioxidants offer the possibility of treatment of OS after ICH. However, researchers still have a long way to go to apply these natural products for the treatment of ICH more widely in the clinic.
Collapse
Affiliation(s)
| | | | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
64
|
Zhou K, Han J, Wang Y, Xu Y, Zhang Y, Zhu C. The therapeutic potential of bone marrow-derived macrophages in neurological diseases. CNS Neurosci Ther 2022; 28:1942-1952. [PMID: 36066198 PMCID: PMC9627381 DOI: 10.1111/cns.13964] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 02/06/2023] Open
Abstract
Circulating monocytes are precursors of both tissue macrophages and dendritic cells, and they can infiltrate the central nervous system (CNS) where they transform into bone marrow-derived macrophages (BMDMs). BMDMs play essential roles in various CNS diseases, thus modulating BMDMs might be a way to treat these disorders because there are currently no efficient therapeutic methods available for most of these neurological diseases. Moreover, BMDMs can serve as promising gene delivery vehicles following bone marrow transplantation for otherwise incurable genetic CNS diseases. Understanding the distinct roles that BMDMs play in CNS diseases and their potential as gene delivery vehicles may provide new insights and opportunities for using BMDMs as therapeutic targets or delivery vehicles. This review attempts to comprehensively summarize the neurological diseases that might be treated by modulating BMDMs or by delivering gene therapies via BMDMs after bone marrow transplantation.
Collapse
Affiliation(s)
- Kai Zhou
- Henan Neurodevelopment Engineering Research Center for ChildrenChildren's Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Jinming Han
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for ChildrenChildren's Hospital Affiliated to Zhengzhou UniversityZhengzhouChina,Department of Hematology and OncologyChildren's Hospital Affiliated to Zhengzhou University, Henan, Children's Hospital, Zhengzhou Children's HospitalZhengzhouChina
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThe Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou UniversityZhengzhouChina
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for ChildrenChildren's Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThe Third Affiliated Hospital and Institute of Neuroscience, Zhengzhou UniversityZhengzhouChina,Centre for Brain Repair and RehabilitationInstitute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
65
|
Chen W, Zhang Y, Zhai X, Xie L, Guo Y, Chen C, Li Y, Wang F, Zhu Z, Zheng L, Wan J, Li P. Microglial phagocytosis and regulatory mechanisms after stroke. J Cereb Blood Flow Metab 2022; 42:1579-1596. [PMID: 35491825 PMCID: PMC9441720 DOI: 10.1177/0271678x221098841] [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] [Indexed: 12/19/2022]
Abstract
Stroke, including ischemic stroke and hemorrhagic stroke can cause massive neuronal death and disruption of brain structure, which is followed by secondary inflammatory injury initiated by pro-inflammatory molecules and cellular debris. Phagocytic clearance of cellular debris by microglia, the brain's scavenger cells, is pivotal for neuroinflammation resolution and neurorestoration. However, microglia can also exacerbate neuronal loss by phagocytosing stressed-but-viable neurons in the penumbra, thereby expanding the injury area and hindering neurofunctional recovery. Microglia constantly patrol the central nervous system using their processes to scour the cellular environment and start or cease the phagocytosis progress depending on the "eat me" or "don't eat me'' signals on cellular surface. An optimal immune response requires a delicate balance between different phenotypic states to regulate neuro-inflammation and facilitate reconstruction after stroke. Here, we examine the literature and discuss the molecular mechanisms and cellular pathways regulating microglial phagocytosis, their resulting effects in brain injury and neural regeneration, as well as the potential therapeutic targets that might modulate microglial phagocytic activity to improve neurological function after stroke.
Collapse
Affiliation(s)
- Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaozhu Zhai
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lv Xie
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunlu Guo
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fajun Wang
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ziyu Zhu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zheng
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqing Wan
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
66
|
Yu F, Wang Y, Stetler AR, Leak RK, Hu X, Chen J. Phagocytic microglia and macrophages in brain injury and repair. CNS Neurosci Ther 2022; 28:1279-1293. [PMID: 35751629 PMCID: PMC9344092 DOI: 10.1111/cns.13899] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/31/2022] [Accepted: 06/04/2022] [Indexed: 12/21/2022] Open
Abstract
AIMS Phagocytosis is the cellular digestion of extracellular particles, such as pathogens and dying cells, and is a key element in the evolution of central nervous system (CNS) disorders. Microglia and macrophages are the professional phagocytes of the CNS. By clearing toxic cellular debris and reshaping the extracellular matrix, microglia/macrophages help pilot the brain repair and functional recovery process. However, CNS resident and invading immune cells can also magnify tissue damage by igniting runaway inflammation and phagocytosing stressed-but viable-neurons. DISCUSSION Microglia/macrophages help mediate intercellular communication and react quickly to the "find-me" signals expressed by dead/dying neurons. The activated microglia/macrophages then migrate to the injury site to initiate the phagocytic process upon encountering "eat-me" signals on the surfaces of endangered cells. Thus, healthy cells attempt to avoid inappropriate engulfment by expressing "do not-eat-me" signals. Microglia/macrophages also have the capacity to phagocytose immune cells that invade the injured brain (e.g., neutrophils) and to regulate their pro-inflammatory properties. During brain recovery, microglia/macrophages engulf myelin debris, initiate synaptogenesis and neurogenesis, and sculpt a favorable extracellular matrix to support network rewiring, among other favorable roles. Here, we review the multilayered nature of phagocytotic microglia/macrophages, including the molecular and cellular mechanisms that govern microglia/macrophage-induced phagocytosis in acute brain injury, and discuss strategies that tap into the therapeutic potential of this engulfment process. CONCLUSION Identification of biological targets that can temper neuroinflammation after brain injury without hindering the essential phagocytic functions of microglia/macrophages will expedite better medical management of the stroke recovery stage.
Collapse
Affiliation(s)
- Fang Yu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yangfan Wang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anne R. Stetler
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rehana K. Leak
- Graduate School of Pharmaceutical SciencesSchool of Pharmacy, Duquesne UniversityPittsburghPennsylvaniaUSA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jun Chen
- Geriatric Research, Education and Clinical CenterVeterans Affairs Pittsburgh Health Care SystemPittsburghPennsylvaniaUSA
- Pittsburgh Institute of Brain Disorders & Recovery and Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
67
|
Lee MJ, Zhu J, An JH, Lee SE, Kim TY, Oh E, Kang YE, Chung W, Heo JY. A transcriptomic analysis of cerebral microvessels reveals the involvement of Notch1 signaling in endothelial mitochondrial-dysfunction-dependent BBB disruption. Fluids Barriers CNS 2022; 19:64. [PMID: 36028880 PMCID: PMC9414148 DOI: 10.1186/s12987-022-00363-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background Endothelial cells (ECs) in cerebral vessels are considered the primary targets in acute hemorrhagic brain injuries. EC dysfunction can aggravate neuronal injuries by causing secondary inflammatory responses and blood–brain barrier (BBB) disruption. Previous studies have reported that enhancement of mitochondrial function within ECs may reduce BBB disruption and decrease the severity of acute brain injuries. However, the molecular signaling pathways through which enhanced EC mitochondrial function is enhanced to exert this BBB protective effect have not been fully elucidated. Methods To identify signaling pathways involved in linking EC-specific mitochondrial dysfunction and BBB disruption, we first performed RNA sequencing using isolated cerebral vessels from TEKCRIF1 KO mice, a mouse strain that displays EC-specific mitochondrial dysfunction. After identification, we assessed the significance of candidate signaling pathways using an intracerebral hemorrhage (ICH) mouse model. BBB integrity was assessed using an IgG leakage assay, and symptomatic changes were evaluated using behavioral assays. Results Transcriptome analyses of the TEKCRIF1 KO mouse revealed significant changes in Notch1 signaling, a pathway intimately involved in BBB maintenance. We also observed a decrease in Notch1 signaling and expression of the mitochondrial oxidative phosphorylation (OxPhos) complex in the ICH mouse model, which also exhibits BBB disruption. To further assess the function of Notch1 signaling in relation to BBB disruption, we injected ICH model mice with adropin, a protein that interacts with the Notch1 ligand NB-3 and activates Notch1 signaling. We found that adropin prevented BBB disruption and reduced the extent (area) of the injury compared with that in vehicle controls, in association with alteration of mitochondrial function. Conclusion These results suggest that the Notch1 signaling pathway acts as an upstream regulator of DEGs and can be a target to regulate the changes involved with endothelial mitochondrial dysfunction-dependent BBB disruption. Thus, treatment methods that activate Notch1 may be beneficial in acute brain injuries by protecting BBB integrity. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00363-7.
Collapse
Affiliation(s)
- Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jiebo Zhu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jong Hun An
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Seong Eun Lee
- Research Center for Endocrine and Metabolic Disease, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University School of Medicine, Deajeon, 35015, Republic of Korea
| | - Tae Yeon Kim
- Bio-Synergy Research Center, Daejeon, 34141, Republic of Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Yea Eun Kang
- Research Center for Endocrine and Metabolic Disease, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University School of Medicine, Deajeon, 35015, Republic of Korea.
| | - Woosuk Chung
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea. .,Department of Anesthesiology and Pain Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea.
| | - Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea. .,Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon, 35015, Republic of Korea. .,Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
68
|
Kosoy R, Fullard JF, Zeng B, Bendl J, Dong P, Rahman S, Kleopoulos SP, Shao Z, Girdhar K, Humphrey J, de Paiva Lopes K, Charney AW, Kopell BH, Raj T, Bennett D, Kellner CP, Haroutunian V, Hoffman GE, Roussos P. Genetics of the human microglia regulome refines Alzheimer's disease risk loci. Nat Genet 2022; 54:1145-1154. [PMID: 35931864 PMCID: PMC9388367 DOI: 10.1038/s41588-022-01149-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/08/2022] [Indexed: 02/07/2023]
Abstract
Microglia are brain myeloid cells that play a critical role in neuroimmunity and the etiology of Alzheimer's disease (AD), yet our understanding of how the genetic regulatory landscape controls microglial function and contributes to AD is limited. Here, we performed transcriptome and chromatin accessibility profiling in primary human microglia from 150 donors to identify genetically driven variation and cell-specific enhancer-promoter (E-P) interactions. Integrative fine-mapping analysis identified putative regulatory mechanisms for 21 AD risk loci, of which 18 were refined to a single gene, including 3 new candidate risk genes (KCNN4, FIBP and LRRC25). Transcription factor regulatory networks captured AD risk variation and identified SPI1 as a key putative regulator of microglia expression and AD risk. This comprehensive resource capturing variation in the human microglia regulome provides insights into the etiology of neurodegenerative disease.
Collapse
Affiliation(s)
- Roman Kosoy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Pengfei Dong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Samir Rahman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Steven P Kleopoulos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Zhiping Shao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jack Humphrey
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katia de Paiva Lopes
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Alexander W Charney
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Brian H Kopell
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Towfique Raj
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
- Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
- Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
69
|
Li C, Wang Y, Xing Y, Han J, Zhang Y, Zhang A, Hu J, Hua Y, Bai Y. Regulation of microglia phagocytosis and potential involvement of exercise. Front Cell Neurosci 2022; 16:953534. [PMID: 35959472 PMCID: PMC9357882 DOI: 10.3389/fncel.2022.953534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/29/2022] [Indexed: 12/11/2022] Open
Abstract
Microglia are considered the main phagocytic cells in the central nervous system, remodeling neural circuits by pruning synapses during development. Microglial phagocytosis is also a crucial process in maintaining adult brain homeostasis and clearing potential toxic factors, which are recognized to be associated with neurodegenerative and neuroinflammatory disorders. For example, microglia can engulf amyloid-β plaques, myelin debris, apoptotic cells, and extracellular harmful substances by expressing a variety of specific receptors on the cell surface or by reprogramming intracellular glucose and lipid metabolism processes. Furthermore, physical exercise has been implicated to be one of the non-pharmaceutical treatments for various nervous system diseases, which is closely related to neuroplasticity and microglia functions including proliferation, activation, and phagocytosis. This review focuses on the central regulatory mechanisms related to microglia phagocytosis and the potential role of exercise training in this process.
Collapse
Affiliation(s)
- Congqin Li
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Wang
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Ying Xing
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Han
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, Brain Science Collaborative Innovation Center, School of Basic Medical Sciences, Institutes of Brain Science, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuqian Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Anjing Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Hu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yulong Bai
| |
Collapse
|
70
|
Wei J, Dai S, Pu C, Luo P, Yang Y, Jiang X, Li X, Lin W, Fei Z. Protective role of TLR9-induced macrophage/microglia phagocytosis after experimental intracerebral hemorrhage in mice. CNS Neurosci Ther 2022; 28:1800-1813. [PMID: 35876247 PMCID: PMC9532915 DOI: 10.1111/cns.13919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Intracerebral hemorrhage (ICH) causes devastating morbidity and mortality, and studies have shown that the toxic components of hematomas play key roles in brain damage after ICH. Recent studies have found that TLR9 participates in regulating the phagocytosis of peripheral macrophages. The current study examined the role of TLR9 in macrophage/microglial (M/M) function after ICH. METHODS RAW264.7 (macrophage), BV2 (microglia), and HT22# (neurons) cell lines were transfected with lentivirus for TLR9 overexpression. Whole blood from C57BL/6 or EGFPTg/+ mice was infused for phagocytosis and injury experiments, and brusatol was used for the experiments. Intraperitoneal injection of the TLR9 agonist ODN1826 or control ODN2138 was performed on days 1, 3, 5, 7, and 28 after ICH to study the effects of TLR9 in mice. In addition, clodronate was coinjected in M/M elimination experiments. The brains were collected for histological and protein experiments at different time points after ICH induction. Cellular and histological methods were used to measure hematoma/iron residual, M/Ms variation, neural injury, and brain tissue loss. Behavioral tests were performed premodeling and on days 1, 3, 7, and 28 post-ICH. RESULTS Overexpression of TLR9 facilitated M/M phagocytosis and protected neurons from blood-derived hazards in vitro. Furthermore, ODN1826 boosted M/M activation and phagocytic function, facilitated hematoma/iron resolution, reduced brain injury, and improved neurological function recovery in ICH mice, which were abolished by clodronate injection. The experimental results indicated that the Nrf2/CD204 pathway participated in TLR9-induced M/M phagocytosis after ICH. CONCLUSION Our study suggests a protective role for TLR9-enhanced M/M phagocytosis via the Nrf2/CD204 pathway after ICH. Our findings may serve as potential targets for ICH treatment.
Collapse
Affiliation(s)
- Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Health Service, Fourth Military Medical University, Xi'an, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen Pu
- Department of Health Service, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuefan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Lin
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
71
|
Investigation on the potential targets of Astragaloside IV against intracerebral hemorrhage based on network pharmacology and experimental validation. Bioorg Chem 2022; 127:105975. [PMID: 35728292 DOI: 10.1016/j.bioorg.2022.105975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022]
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening type of stroke that affects millions of individuals worldwide. Astragaloside IV (AS-IV), the main bioactive ingredient in Radix Astragali, has been linked to a variety of pharmacologic actions, including stroke. However, the effects and potential mechanisms of AS-IV on hematoma absorption after ICH are still unknown. The study aims to identify potential targets and regulation mechanisms of AS-IV on hematoma absorption after ICH. Network pharmacology, molecular docking, pharmacodynamic study, and western blot were used in this study to explore the potential mechanisms. The results showed that AS-IV could improve the hematoma absorption and neurological outcomes in collagenase VII induced rat ICH models. Molecular docking results had shown that PI3K and AKT were the potential targets of AS-IV against ICH. The experimental validation showed that AS-IV could reduce phosphorylation expression of PI3K and AKT, thereby inhibiting the NF-κB and increasing CD36 expression. This study demonstrated that AS-IV could play a critical role on hematoma absorption after ICH by regulating the PI3K/AKT signaling pathway and promoting CD36 phagocytosis, which provided a new thought for the drug development of ICH.
Collapse
|
72
|
Shen Q, Zhao L, Pan L, Li D, Chen G, Chen Z, Jiang Z. Soluble SIRP-Alpha Promotes Murine Acute Lung Injury Through Suppressing Macrophage Phagocytosis. Front Immunol 2022; 13:865579. [PMID: 35634325 PMCID: PMC9133620 DOI: 10.3389/fimmu.2022.865579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/13/2022] [Indexed: 11/17/2022] Open
Abstract
Soluble signal regulatory protein-alpha (SIRP-alpha) is elevated in bronchoalveolar lavage (BAL) of mice with lipopolysaccharides (LPS)-induced acute lung injury (ALI). To define the role of soluble SIRP-alpha in the pathogenesis of ALI, we established murine ALI in wild-type (WT) and SIRP-alpha knock-out (KO) mice by intratracheal administration of LPS. The results indicated that lack of SIRP-alpha significantly reduced the pathogenesis of ALI, in association with attenuated lung inflammation, infiltration of neutrophils and expression of pro-inflammatory cytokines in mice. In addition, lack of SIRP-alpha reduced the expression of pro-inflammatory cytokines in LPS-treated bone marrow-derived macrophages (BMDMs) from KO mice, accompanied with improved macrophage phagocytosis. Blockade of soluble SIRP-alpha activity in ALI BAL by anti-SIRP-alpha antibody (aSIRP) effectively reduced the expression of TNF-alpha and IL-6 mRNA transcripts and proteins, improved macrophage phagocytosis in vitro. In addition, lack of SIRP-alpha reduced activation of Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP-1) and improved activation of signal transducer and activator of transcription-3 (STAT3) and STAT6. Suppression of SHP-1 activity by tyrosine phosphatase inhibitor 1 (TPI-1) increased activation of STAT3 and STAT6, and improved macrophage phagocytosis, that was effectively reversed by STAT3 and STAT6 inhibitors. Thereby, SIRP-alpha suppressed macrophage phagocytosis through activation of SHP-1, subsequently inhibiting downstream STAT3 and STAT6 signaling. Lack of SIRP-alpha attenuated murine ALI possibly through increasing phagocytosis, and improving STAT3 and STAT6 signaling in macrophages. SIRP-alpha would be promising biomarker and molecular target in the treatment of murine ALI and patients with acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Qinjun Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Zhao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linyue Pan
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dandan Li
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
73
|
Zhang L, Zhou H, Wang S, Guan Y, Zhang C, Fang D. Changes in microglia during drug treatment of stroke. IBRAIN 2022; 8:227-240. [PMID: 37786889 PMCID: PMC10528798 DOI: 10.1002/ibra.12037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/07/2022] [Accepted: 04/20/2022] [Indexed: 10/04/2023]
Abstract
Microglia are the main immune cells in the brain and the first defense barrier of the nervous system. Microglia play a complex role in the process of stroke. A growing number of studies focus on the mechanism of action of drugs functions and how to regulate microglia. Therefore, we talk about the pathophysiological mechanisms of stroke and elaborate on the microglia signaling pathways of drug action in stroke models and how these drugs play a role in stroke treatment in this review. Understanding how drugs modulate proinflammatory and anti-inflammatory responses of microglia may be critical to implementing therapeutic strategies using immune interventions in stroke.
Collapse
Affiliation(s)
- Ling‐Jing Zhang
- Department of AnesthesiaZunyi Medical UniversityZunyiGuizhouChina
| | - Hong‐Su Zhou
- Department of AnesthesiaGraduate School of Zunyi Medical UniversityZunyiGuizhouChina
| | - Shi‐Ya Wang
- Department of AnesthesiaZunyi Medical UniversityZunyiGuizhouChina
| | - Yi‐Huan Guan
- Department of AnesthesiaZunyi Medical UniversityZunyiGuizhouChina
| | - Chao Zhang
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - De‐Rong Fang
- Department of Family PlanningAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
74
|
Liu J, Li N, Zhu Z, Kiang KMY, Ng ACK, Dong CM, Leung GKK. Vitamin D Enhances Hematoma Clearance and Neurologic Recovery in Intracerebral Hemorrhage. Stroke 2022; 53:2058-2068. [PMID: 35514286 DOI: 10.1161/strokeaha.121.037769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Erythrophagocytosis by reparative monocyte-derived macrophage contributes to hematoma clearance and neurological recovery after intracerebral hemorrhage (ICH). Vitamin D (VitD) is a neuroprotective hormone and regulates the differentiation of monocyte-derived macrophage from monocytes. In this study, we examined the effects of VitD supplementation on monocyte-derived macrophage and hematoma clearance in rodent with ICH. METHODS Neurobehavioral functions and hematoma volume were assessed using a collagenase injection model in both young- and middle-aged mice with or without VitD treatment given 2 hours post-ICH induction. We used flow cytometry to analyze CD36 expression and macrophage and undifferentiated monocyte cell numbers during in vivo erythrophagocytosis in collagenase and autologous blood injection models. Western blot analysis and immunofluorescence were used to assess the expression levels of the PPAR-γ (peroxisome proliferator-activated receptor γ)-CD36 axis and CD206. A macrophage differentiation study was conducted on murine bone marrow-derived monocytes. RESULTS VitD promoted neurological recovery and facilitated hematoma clearance in both young- and middle-aged mice after ICH. Within the perihematomal region, mature macrophages, rather than undifferentiated monocytes, expressed higher levels of CD36 in driving erythrocyte clearance. VitD increased the macrophage number but decreased the monocyte number and elevated the levels of CD36 and PPAR-γ in the brain. In vitro, VitD accelerated the differentiation of reparative macrophages from bone marrow-derived monocytes. CONCLUSIONS VitD promotes reparative macrophage differentiation, facilitates hematoma clearance, and improves neurobehavioral performance in mice with ICH, suggesting that VitD should be further examined as a potentially promising treatment for ICH.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, China (J.L., N.L., Z.Z., K.K., A.N.)
| | - Ning Li
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, China (J.L., N.L., Z.Z., K.K., A.N.).,Department of Neurosurgery, Zhongda Hospital, Southeast University, Nanjing, China (N.L.)
| | - Zhiyuan Zhu
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, China (J.L., N.L., Z.Z., K.K., A.N.).,Department of Functional neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China (Z.Z.)
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, China (J.L., N.L., Z.Z., K.K., A.N.)
| | - Anson Cho Kiu Ng
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, China (J.L., N.L., Z.Z., K.K., A.N.)
| | - Celia M Dong
- Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, China (C.M.D.)
| | | |
Collapse
|
75
|
Abstract
Stroke remains a significant unmet clinical need with few treatment options that have a very narrow therapeutic window, thereby causing massive mortality and morbidity in the United States and around the world. Accordingly, finding safe and effective novel treatments with a wider therapeutic window stands as an urgent need in stroke. The progressive inflammation that occurs centrally and peripherally after stroke serves as a unique therapeutic target to retard and even halt the secondary cell death. Stem cell therapy represents a potent approach that can diminish inflammation in both the stroke brain and periphery (eg, spleen), advancing a paradigm shift from a traditionally brain-focused therapy to treating stroke as a neurological disorder with a significant peripheral pathology. The purpose of this review article is to highlight the inflammation-mediated secondary cell death that plagues both brain and spleen in stroke and to evaluate the therapeutic potential of stem cell therapy in dampening these inflammatory responses.
Collapse
Affiliation(s)
- Stefan Anthony
- Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL 34211, USA
| | - Dorothy Cabantan
- Michigan State University College of Osteopathic Medicine, 965 Wilson Rd, East Lansing, MI 48824, USA
| | - Molly Monsour
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
76
|
Magid-Bernstein J, Girard R, Polster S, Srinath A, Romanos S, Awad IA, Sansing LH. Cerebral Hemorrhage: Pathophysiology, Treatment, and Future Directions. Circ Res 2022; 130:1204-1229. [PMID: 35420918 PMCID: PMC10032582 DOI: 10.1161/circresaha.121.319949] [Citation(s) in RCA: 168] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high morbidity and mortality. This review article focuses on the epidemiology, cause, mechanisms of injury, current treatment strategies, and future research directions of ICH. Incidence of hemorrhagic stroke has increased worldwide over the past 40 years, with shifts in the cause over time as hypertension management has improved and anticoagulant use has increased. Preclinical and clinical trials have elucidated the underlying ICH cause and mechanisms of injury from ICH including the complex interaction between edema, inflammation, iron-induced injury, and oxidative stress. Several trials have investigated optimal medical and surgical management of ICH without clear improvement in survival and functional outcomes. Ongoing research into novel approaches for ICH management provide hope for reducing the devastating effect of this disease in the future. Areas of promise in ICH therapy include prognostic biomarkers and primary prevention based on disease pathobiology, ultra-early hemostatic therapy, minimally invasive surgery, and perihematomal protection against inflammatory brain injury.
Collapse
Affiliation(s)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
77
|
Kutchy NA, Ma R, Liu Y, Buch S, Hu G. Extracellular Vesicle-Mediated Delivery of Ultrasmall Superparamagnetic Iron Oxide Nanoparticles to Mice Brain. Front Pharmacol 2022; 13:819516. [PMID: 35462907 PMCID: PMC9022024 DOI: 10.3389/fphar.2022.819516] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are small lipid membrane-bound vesicles that can pass the blood–brain barrier. Therefore, EVs could be used for the delivery of therapeutics to the brain. Herein, we investigated the biodistribution of intranasal perfusion of ultrasmall superparamagnetic iron oxide (USPIO)-labeled astrocyte-derived EVs (ADEVs) in mice. We used Western blotting, transmission electron microscopy (TEM), and nanoparticle uptake assay to characterize ADEVs. In addition, intranasal perfusion coupled with magnetic resonance imaging (MRI) was employed to determine the distribution of USPIO-labeled ADEVs in mice. Our results showed the uptake of USPIO by mouse astrocytes and ADEVs. In addition, we confirmed the biodistribution of ADEVs in the brain and other internal organs, including the kidneys, liver, and spleen. Our results suggest that USPIO did not affect mouse astrocyte cell survivability and EV release. Therefore, intranasal delivery of therapeutic loaded EVs could be used for the treatment of various brain disorders.
Collapse
Affiliation(s)
- Naseer A. Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Anatomy, Physiology, and Pharmacology, School of Veterinary Medicine, St. George’s University, St. George’s, Grenada
| | - Rong Ma
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yutong Liu
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Guoku Hu,
| |
Collapse
|
78
|
Soluble Trem2 is a negative regulator of erythrophagocytosis after intracerebral hemorrhage in a CD36 receptor recycling manner. J Adv Res 2022; 44:185-199. [PMID: 36725189 PMCID: PMC9936424 DOI: 10.1016/j.jare.2022.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/23/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Microglia and macrophages participate in hematoma clearance after intracerebral hemorrhage (ICH), thereby facilitating tissue restoration and neurological recovery. Triggering receptor expressed on myeloid cells 2 (Trem2) has been indicated as a major pathology-induced immune signaling hub on the microglial/macrophage surface. Soluble Trem2 (sTrem2), the proteolytic form of Trem2, is abundant in the body fluid and is positively correlated with the pathological process. OBJECTIVES In the present study, we aimed to investigate the potential role of sTrem2 in hematoma resolution after ICH and to elucidate its underlying mechanisms. METHODS We explored the biological functions of sTrem2 in the murine ICH brain by stereotaxic injection of recombinant sTrem2 protein or by adeno-associated virus-mediated expression. Erythrocyte phagocytosis was assessed using flow cytometry and immunofluorescence. Western blotting was performed to evaluate protein expression. Changes in behavior, sTrem2-induced down-stream pathway, and microglia were examined. RESULTS sTrem2 impedes hematoma resolution and impairs functional motor and sensory recovery. Interestingly, sTrem2 bypasses full-length Trem2, negatively regulating microglial/macrophage erythrophagocytosis, and promotes an inflammatory phenotype, which is associated with reduced retromer levels and impaired recycling of the pro-erythrophagocytic receptor CD36. Rescue of retromer Vps35 abolishes the phagocytosis-inhibiting effects and lysosome-dependent CD36 degradation caused by sTrem2. CONCLUSION These findings indicate sTrem2 as a negative factor against microglia/macrophage-mediated hematoma and related neuronal damage clearance, provide insight into the mechanisms by which erythrophagocytosis is regulated and how it may be impaired after ICH, and suggest that the anti-proteolytic activity of Trem2 can be explored for ICH therapy.
Collapse
|
79
|
Liu J, Zhu Z, Leung GKK. Erythrophagocytosis by Microglia/Macrophage in Intracerebral Hemorrhage: From Mechanisms to Translation. Front Cell Neurosci 2022; 16:818602. [PMID: 35237132 PMCID: PMC8882619 DOI: 10.3389/fncel.2022.818602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition characterized by hematoma related mass effect. Microglia/macrophage (M φ) are rapidly recruited in order to remove the red blood cells through erythrophagocytosis. Efficient erythrophagocytosis can detoxify hemolytic products and facilitate neurological recovery after ICH. The underlying mechanisms include modulation of inflammatory response and oxidative stress, among others. It is a dynamic process mediated by a cascade of signal transduction, including “find-me” signals, “eat-me” signals and a set of phagocytotic receptors-ligand pairs that may be exploited as therapeutic targets. This review summarizes mechanistic signaling pathways of erythrophagocytosis and highlights the potential of harnessing M φ-mediated phagocytosis for ICH treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Zhiyuan Zhu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- *Correspondence: Gilberto Ka-Kit Leung,
| |
Collapse
|
80
|
Tsai HH, Hsieh YC, Lin JS, Kuo ZT, Ho CY, Chen CH, Chang CF. Functional Investigation of Meningeal Lymphatic System in Experimental Intracerebral Hemorrhage. Stroke 2022; 53:987-998. [DOI: 10.1161/strokeaha.121.037834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Promotion of hematoma resolution in a timely manner reduces intracerebral hemorrhage (ICH) brain injury induced by toxic blood components and subsequent neuroinflammation. The meningeal lymphatic system is responsible for clearance of macromolecules and pathogenic substances from the central nervous system; however, its role in intraparenchymal hematoma clearance and ICH outcomes is unknown. In the present study, we aimed to understand the contribution of the meningeal lymphatic system to ICH pathologies and to test whether pharmacological enhancement of meningeal lymphatic function promotes hematoma resolution and brain recovery after ICH.
Methods:
Immunofluorescence of whole-mount meninges was used to measure complexity and coverage level of meningeal lymphatic vasculature following ICH induction. Fluorescent microbeads and PKH-26-labeled erythrocytes were used to evaluate drainage function of the meningeal lymphatic system. Visudyne treatment, deep cervical lymph node ligation, and VEGF (vascular endothelial growth factor)-C injection were performed to manipulate meningeal lymphatic function. Neurobehavioral performance and hematoma volume were assayed by the cylinder test and histological measurements. Iron deposition, residual erythrocytes, neuronal loss, and astrogliosis were assessed by immunohistochemistry and antibody-based fluorescence staining.
Results:
Meningeal lymphangiogenesis and enhanced lymphatic drainage occurred during the late phase of ICH. Ablation and blockage of meningeal lymphatic vessels impeded hematoma clearance, whereas pharmacological enhancement of their function reduced hematoma volume, improved behavioral performance, and reduced brain residual erythrocytes, iron deposition, neuronal loss, and astroglial activation.
Conclusions:
Early enhancement of meningeal lymphatic function is beneficial for ICH recovery. Targeting the meningeal lymphatic system is therefore a potential therapeutic approach for treating ICH.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei (H.-H.T.)
- Department of Neurology, National Taiwan University Hospital, Taipei (H.-H.T.)
| | - Yung-Chia Hsieh
- School of Medicine, National Taiwan University College of Medicine, Taipei. (Y.-C.H.)
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Jhih Syuan Lin
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Zi-Ting Kuo
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Chi-Ying Ho
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Chih-Hung Chen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| |
Collapse
|
81
|
Novel targets, treatments, and advanced models for intracerebral haemorrhage. EBioMedicine 2022; 76:103880. [PMID: 35158309 PMCID: PMC8850756 DOI: 10.1016/j.ebiom.2022.103880] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/17/2022] [Accepted: 01/28/2022] [Indexed: 12/20/2022] Open
Abstract
Intracerebral haemorrhage (ICH) is the second most common type of stroke and a major cause of mortality and disability worldwide. Despite advances in surgical interventions and acute ICH management, there is currently no effective therapy to improve functional outcomes in patients. Recently, there has been tremendous progress uncovering new pathophysiological mechanisms underlying ICH that may pave the way for the development of therapeutic interventions. Here, we highlight emerging targets, but also existing gaps in preclinical animal modelling that prevent their exploitation. We particularly focus on (1) ICH aetiology, (2) the haematoma, (3) inflammation, and (4) post-ICH pathology. It is important to recognize that beyond neurons and the brain, other cell types and organs are crucially involved in ICH pathophysiology and successful interventions likely will need to address the entire organism. This review will spur the development of successful therapeutic interventions for ICH and advanced animal models that better reflect its aetiology and pathophysiology.
Collapse
|
82
|
Pan R, Yu S, Zhang H, Timmins GS, Weaver J, Yang Y, Zhou X, Liu KJ. Endogenous zinc protoporphyrin formation critically contributes to hemorrhagic stroke-induced brain damage. J Cereb Blood Flow Metab 2021; 41:3232-3247. [PMID: 34187233 PMCID: PMC8669275 DOI: 10.1177/0271678x211028475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hemorrhagic stroke is a leading cause of death. The causes of intracerebral hemorrhage (ICH)-induced brain damage are thought to include lysis of red blood cells, hemin release and iron overload. These mechanisms, however, have not proven very amenable to therapeutic intervention, and so other mechanistic targets are being sought. Here we report that accumulation of endogenously formed zinc protoporphyrin (ZnPP) also critically contributes to ICH-induced brain damage. ICH caused a significant accumulation of ZnPP in brain tissue surrounding hematoma, as evidenced by fluorescence microscopy of ZnPP, and further confirmed by fluorescence spectroscopy and supercritical fluid chromatography-mass spectrometry. ZnPP formation was dependent upon both ICH-induced hypoxia and an increase in free zinc accumulation. Notably, inhibiting ferrochelatase, which catalyzes insertion of zinc into protoporphyrin, greatly decreased ICH-induced endogenous ZnPP generation. Moreover, a significant decrease in brain damage was observed upon ferrochelatase inhibition, suggesting that endogenous ZnPP contributes to the damage in ICH. Our findings reveal a novel mechanism of ICH-induced brain damage through ferrochelatase-mediated formation of ZnPP in ICH tissue. Since ferrochelatase can be readily inhibited by small molecules, such as protein kinase inhibitors, this may provide a promising new and druggable target for ICH therapy.
Collapse
Affiliation(s)
- Rong Pan
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Song Yu
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Haikun Zhang
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Graham S Timmins
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - John Weaver
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Yirong Yang
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, USA
| |
Collapse
|
83
|
Pu H, Ma C, Zhao Y, Wang Y, Zhang W, Miao W, Yu F, Hu X, Shi Y, Leak RK, Hitchens TK, Dixon CE, Bennett MV, Chen J. Intranasal delivery of interleukin-4 attenuates chronic cognitive deficits via beneficial microglial responses in experimental traumatic brain injury. J Cereb Blood Flow Metab 2021; 41:2870-2886. [PMID: 34259069 PMCID: PMC8545055 DOI: 10.1177/0271678x211028680] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Traumatic brain injury (TBI) is commonly followed by long-term cognitive deficits that severely impact the quality of life in survivors. Recent studies suggest that microglial/macrophage (Mi/MΦ) polarization could have multidimensional impacts on post-TBI neurological outcomes. Here, we report that repetitive intranasal delivery of interleukin-4 (IL-4) nanoparticles for 4 weeks after controlled cortical impact improved hippocampus-dependent spatial and non-spatial cognitive functions in adult C57BL6 mice, as assessed by a battery of neurobehavioral tests for up to 5 weeks after TBI. IL-4-elicited enhancement of cognitive functions was associated with improvements in the integrity of the hippocampus at the functional (e.g., long-term potentiation) and structural levels (CA3 neuronal loss, diffusion tensor imaging of white matter tracts, etc.). Mechanistically, IL-4 increased the expression of PPARγ and arginase-1 within Mi/MΦ, thereby driving microglia toward a global inflammation-resolving phenotype. Notably, IL-4 failed to shift microglial phenotype after TBI in Mi/MΦ-specific PPARγ knockout (mKO) mice, indicating an obligatory role for PPARγ in IL-4-induced Mi/MΦ polarization. Accordingly, post-TBI treatment with IL-4 failed to improve hippocampal integrity or cognitive functions in PPARγ mKO mice. These results demonstrate that administration of exogenous IL-4 nanoparticles stimulates PPARγ-dependent beneficial Mi/MΦ responses, and improves hippocampal function after TBI.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cheng Ma
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yongfang Zhao
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yangfan Wang
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wenting Zhang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wanying Miao
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fang Yu
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yejie Shi
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rehana K Leak
- Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - T Kevin Hitchens
- Animal Imaging Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Vl Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
84
|
Ge JW, Deng SJ, Xue ZW, Liu PY, Yu LJ, Li JN, Xia SN, Gu Y, Bao XY, Lan Z, Xu Y, Zhu XL. Imperatorin inhibits mitogen-activated protein kinase and nuclear factor kappa-B signaling pathways and alleviates neuroinflammation in ischemic stroke. CNS Neurosci Ther 2021; 28:116-125. [PMID: 34674376 PMCID: PMC8673701 DOI: 10.1111/cns.13748] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 09/29/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
AIMS Microglia-mediated neuroinflammation plays an important role in the pathological process of ischemic stroke, and the effect of imperatorin on post-stroke neuroinflammation is not fully understood. METHODS Primary microglia were treated with imperatorin for 2 h followed by LPS (100 ng/ml) for 24 h. The expression of inflammatory cytokines was detected by RT-PCR, ELISA, and Western blot. The activation of MAPK and NF-κB signaling pathways were analyzed by Western blot. The ischemic insult was determined using a transient middle cerebral artery occlusion (tMCAO) model in C57BL/6J mice. Behavior tests were used to assess the neurological deficits of MCAO mice. TTC staining was applied to measure infract volume. RESULTS Imperatorin suppressed LPS-induced activation of microglia and pro-inflammatory cytokines release and attenuated ischemic injury in MCAO mice. The results of transcriptome sequencing and Western blot revealed that downregulation of MAPK and NF-κB pathways might contribute to the protective effects of imperatorin. CONCLUSIONS Imperatorin downregulated MAPK and NF-κB signaling pathways and exerted anti-inflammatory effects in ischemic stroke, which indicated that imperatorin might be a potential compound for the treatment of stroke.
Collapse
Affiliation(s)
- Jian-Wei Ge
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Shi-Ji Deng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Zhi-Wei Xue
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Pin-Yi Liu
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Lin-Jie Yu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Jiang-Nan Li
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Sheng-Nan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Yue Gu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Xin-Yu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China
| | - Zhen Lan
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China.,Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xiao-Lei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, PR China.,Institute of Brain Sciences, Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, PR China.,Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, PR China.,Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, PR China.,Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| |
Collapse
|
85
|
Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2021; 42:2505-2525. [PMID: 34460037 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
|
86
|
Wang J, Tang XQ, Xia M, Li CC, Guo C, Ge HF, Yin Y, Wang B, Chen WX, Feng H. Iron chelation suppresses secondary bleeding after intracerebral hemorrhage in angiotensin II-infused mice. CNS Neurosci Ther 2021; 27:1327-1338. [PMID: 34346561 PMCID: PMC8504530 DOI: 10.1111/cns.13706] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 12/01/2022] Open
Abstract
AIMS Secondary bleeding and further hematoma expansion (HE) aggravate brain injury after intracerebral hemorrhage (ICH). The majority of HE results from hypertensive ICH. Previous study reported higher iron content in the brains of hypertensive patients. Iron overload exacerbates the risk of hemorrhagic transformation in thromboembolic stroke mice. Whether iron overload during the process of hypertension participates in secondary bleeding of hypertensive ICH remains unclear. METHODS Hypertension was induced by continuous infusion of angiotensin II (Ang II) with an osmotic pump into C57BL/6 mice. ICH was simulated by intrastriatal injection of the liquid polymer Onyx-18. Iron chelation and iron overload was achieved by deferoxamine mesylate or iron dextran injection. Secondary bleeding was quantified by measuring the hemoglobin content in the ipsilateral brain hemisphere. RESULTS Ang II-induced hypertensive mice showed increased iron accumulation in the brain and expanded secondary hemorrhage after ICH modeling. Moreover, iron chelation suppressed while iron overload aggravated secondary bleeding. Mechanistically, iron exacerbated the loss of contractile cerebral vascular smooth muscle cells (VSMCs), aggravated blood-brain barrier (BBB) leakage in Ang II-induced hypertensive mice, and increased glial and MMP9 accumulation after ICH. CONCLUSION Iron overload plays a key role in secondary bleeding after ICH in Ang II-induced hypertensive mice. Iron chelation during the process of Ang II-induced hypertension suppresses secondary bleeding after ICH.
Collapse
Affiliation(s)
- Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiao-Qin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Cheng-Cheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Hong-Fei Ge
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Bo Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Wei-Xiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
87
|
Role of Interleukin-1 Receptor-Like 1 (ST2) in Cerebrovascular Disease. Neurocrit Care 2021; 35:887-893. [PMID: 34231185 DOI: 10.1007/s12028-021-01284-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022]
Abstract
Following both ischemic and hemorrhagic stroke, innate immune cells initiate a proinflammatory response that further exacerbate tissue injury in the acute phase, but these cells also play an important reparative role thereafter. Numerous cytokines and signaling pathways have been implicated in driving the deleterious proinflammatory response, but less is known about the mediators that connect the initial vascular injury to the systemic immune response and the relationship between proinflammatory and reparative immune responses. The Interleukin-33 (IL-33) and serum stimulation-2 (ST2) axis is an interleukin signaling pathway that is a prime candidate to fulfill this role. In this review, we describe the biology of the IL-33/ST2 system, present evidence that its soluble decoy receptor, soluble ST2 (sST2), plays a key role in secondary neurologic injury after stroke, and discuss this in the context of the known role of IL-33/ST2 in other disease.
Collapse
|
88
|
Kurosaka M, Ogura Y, Sato S, Kohda K, Funabashi T. Transcription factor signal transducer and activator of transcription 6 (STAT6) is an inhibitory factor for adult myogenesis. Skelet Muscle 2021; 11:14. [PMID: 34051858 PMCID: PMC8164270 DOI: 10.1186/s13395-021-00271-8] [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: 11/17/2020] [Accepted: 05/18/2021] [Indexed: 01/25/2023] Open
Abstract
Background The signal transducer and activator of transcription 6 (STAT6) transcription factor plays a vitally important role in immune cells, where it is activated mainly by interleukin-4 (IL-4). Because IL-4 is an essential cytokine for myotube formation, STAT6 might also be involved in myogenesis as part of IL-4 signaling. This study was conducted to elucidate the role of STAT6 in adult myogenesis in vitro and in vivo. Methods Myoblasts were isolated from male mice and were differentiated on a culture dish to evaluate the change in STAT6 during myotube formation. Then, the effects of STAT6 overexpression and inhibition on proliferation, differentiation, and fusion in those cells were studied. Additionally, to elucidate the myogenic role of STAT6 in vivo, muscle regeneration after injury was evaluated in STAT6 knockout mice. Results IL-4 can increase STAT6 phosphorylation, but STAT6 phosphorylation decreased during myotube formation in culture. STAT6 overexpression decreased, but STAT6 knockdown increased the differentiation index and the fusion index. Results indicate that STAT6 inhibited myogenin protein expression. Results of in vivo experiments show that STAT6 knockout mice exhibited better regeneration than wild-type mice 5 days after cardiotoxin-induced injury. It is particularly interesting that results obtained using cells from STAT6 knockout mice suggest that this STAT6 inhibitory action for myogenesis was not mediated by IL-4 but might instead be associated with p38 mitogen-activated protein kinase phosphorylation. However, STAT6 was not involved in the proliferation of myogenic cells in vitro and in vivo. Conclusion Results suggest that STAT6 functions as an inhibitor of adult myogenesis. Moreover, results suggest that the IL-4-STAT6 signaling axis is unlikely to be responsible for myotube formation. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00271-8.
Collapse
Affiliation(s)
- Mitsutoshi Kurosaka
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Shuichi Sato
- School of Kinesiology, The University of Louisiana at Lafayette, Lafayette, LA, USA.,New Iberia Research Center, The University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Kazuhisa Kohda
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toshiya Funabashi
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
89
|
Lu J, Li Z, Zhao Q, Liu D, Mei YA. Neuritin improves the neurological functional recovery after experimental intracerebral hemorrhage in mice. Neurobiol Dis 2021; 156:105407. [PMID: 34058347 DOI: 10.1016/j.nbd.2021.105407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/06/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide, with intracerebral hemorrhage (ICH) being the most lethal subtype. Neuritin (Nrn) is a neurotropic factor that has been reported to have neuroprotective effects in acute brain and spinal cord injury. However, whether Nrn has a protective role in ICH has not been investigated. In this study, ICH was induced in C57BL/6 J mice by injection of collagenase VII, while the overexpression of Nrn in the striatum was induced by an adeno-associated virus serotype 9 (AAV9) vector. We found that compared with GFP-ICH mice, Nrn-ICH mice showed improved performance in the corner, cylinder and forelimb tests after ICH, and showed less weight loss and more rapid weight recovery. Overexpression of Nrn reduced brain lesions, edema, neuronal death and white matter and synaptic integrity dysfunction caused by ICH. Western blot results showed that phosphorylated PERK and ATF4 were significantly inhibited, while phosphorylation of Akt/mammalian target of rapamycin was increased in the Nrn-ICH group, compared with the GFP-ICH group. Whole cell recording from motor neurons indicated that overexpression of Nrn reversed the decrease of spontaneous excitatory postsynaptic currents (sEPSCs) and action potential frequencies induced by ICH. These data show that Nrn improves neurological deficits in mice with ICH by reducing brain lesions and edema, inhibiting neuronal death, and possibly by increasing neuronal connections.
Collapse
Affiliation(s)
- Junmei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Zhaoyang Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qianru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Dongdong Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
90
|
Liu J, Liu L, Wang X, Jiang R, Bai Q, Wang G. Microglia: A Double-Edged Sword in Intracerebral Hemorrhage From Basic Mechanisms to Clinical Research. Front Immunol 2021; 12:675660. [PMID: 34025674 PMCID: PMC8135095 DOI: 10.3389/fimmu.2021.675660] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS). It is well established that microglia are activated and polarized to acquire different inflammatory phenotypes, either pro-inflammatory or anti-inflammatory phenotypes, which act as a critical component in the neuroinflammation following intracerebral hemorrhage (ICH). Microglia produce pro-inflammatory mediators at the early stages after ICH onset, anti-inflammatory microglia with neuroprotective effects appear to be suppressed. Previous research found that driving microglia towards an anti-inflammatory phenotype could restrict inflammation and engulf cellular debris. The principal objective of this review is to analyze the phenotypes and dynamic profiles of microglia as well as their shift in functional response following ICH. The results may further the understanding of the body's self-regulatory functions involving microglia following ICH. On this basis, suggestions for future clinical development and research are provided.
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Lirong Liu
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Xiaoyu Wang
- Xiangya Medical College of Central South University, Changsha, China
| | - Rundong Jiang
- Xiangya Medical College of Central South University, Changsha, China
| | - Qinqin Bai
- Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Gaiqing Wang
- Department of Neurology, Sanya Central Hospital (Hainan Third People's Hospital), Sanya, China
| |
Collapse
|
91
|
Mike JK, Ferriero DM. Efferocytosis Mediated Modulation of Injury after Neonatal Brain Hypoxia-Ischemia. Cells 2021; 10:1025. [PMID: 33925299 PMCID: PMC8146813 DOI: 10.3390/cells10051025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Neonatal brain hypoxia-ischemia (HI) is a leading cause of morbidity and long-term disabilities in children. While we have made significant progress in describing HI mechanisms, the limited therapies currently offered for HI treatment in the clinical setting stress the importance of discovering new targetable pathways. Efferocytosis is an immunoregulatory and homeostatic process of clearance of apoptotic cells (AC) and cellular debris, best described in the brain during neurodevelopment. The therapeutic potential of stimulating defective efferocytosis has been recognized in neurodegenerative diseases. In this review, we will explore the involvement of efferocytosis after a stroke and HI as a promising target for new HI therapies.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Donna Marie Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Neurology Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
92
|
Xu D, Gao Q, Wang F, Peng Q, Wang G, Wei Q, Lei S, Zhao S, Zhang L, Guo F. Sphingosine-1-phosphate receptor 3 is implicated in BBB injury via the CCL2-CCR2 axis following acute intracerebral hemorrhage. CNS Neurosci Ther 2021; 27:674-686. [PMID: 33645008 PMCID: PMC8111497 DOI: 10.1111/cns.13626] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a catastrophic cerebrovascular disease with high morbidity and mortality. Evidence demonstrated that sphingosine-1-phosphate receptor (S1PR) plays a vital role in inflammatory damage via the upregulation of CCL2 expression. However, whether S1PR3 is involved in blood-brain barrier (BBB) breakdown via CCL2 activation after ICH has not been described. METHODS We investigated the expression profiles of all S1PRs using high-throughput RNA-seq analysis and RT-PCR. The potential role of S1PR3 and interaction between S1PR3 and CCL2 were evaluated via Western blotting, immunofluorescence, and flow cytometry. BBB disruption was examined via magnetic resonance imaging, transmission electron microscopy, and Evans blue extravasation. Microglial activation, proliferation, and polarization were assessed via histopathological analysis. The expression levels of CCL2, p-p38 MAPK, ICAM-1, and ZO-1 were examined in vitro and in vivo. RESULTS The present results showed that the levels of S1PR3 and its ligand, sphingosine 1-phosphate (S1P), were dramatically increased following ICH, which regulated the expression of CCL2 and p38MAPK. Moreover, reductions in brain edema volume, amelioration of BBB integrity, and improvements in behavioral deficits were achieved after the administration of CAY10444, an S1PR3 antagonist, to rats. Remarkably increased CCL2, p-p38MAPK, and ICAM-1 expression and decreased ZO-1 expression were observed in cocultured human astrocytes (HAs) and hCMEC/D3 cells after S1P stimulation. However, the expression levels of CCL2, p-p38 MAPK, and ICAM-1 were decreased and ZO-1 expression was increased after S1PR3 inhibition. In addition, microglial proliferation and M1 polarization were attenuated after CAY10444 administration. CONCLUSION To the best of our knowledge, this is the first demonstration of the neuroprotective role of S1PR3 modulation in maintaining BBB integrity by inhibiting the S1PR3-CCL2 axis after ICH, providing a novel treatment for ICH by targeting S1PR3.
Collapse
Affiliation(s)
- Dingkang Xu
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Qiang Gao
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Fang Wang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Qianrui Peng
- Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Guoqing Wang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Qingjie Wei
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Shixiong Lei
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Shengqi Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Longxiao Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| | - Fuyou Guo
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, PR China
| |
Collapse
|