1
|
Jiang M, Xu Y, Luan X, Wu K, Li Z, Xu HE, Zhang S, Jiang Y, Yin W. Structural basis of the cysteinyl leukotriene receptor type 2 activation by LTD4. Proc Natl Acad Sci U S A 2025; 122:e2417148122. [PMID: 40193607 PMCID: PMC12012480 DOI: 10.1073/pnas.2417148122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/18/2025] [Indexed: 04/09/2025] Open
Abstract
The G protein-coupled cysteinyl leukotriene receptor CysLT2R plays intricate roles in the physiology and pathogenesis of inflammation-related processes. It has garnered increasing attention as a potential therapeutic target for atopic asthma, brain injury, central nervous system disorders, and various types of cancer. In this study, we present the cryo-electron microscopy structure of the cysteinyl leukotriene D4 (LTD4)-bound human CysLT2R in complex with a Gαq protein, adopting an active conformation at a resolution of 3.15 Å. The structure elucidates a spacious polar pocket designed to accommodate the two branched negative ends of LTD4 and reveals a lateral ligand access route into the orthosteric pocket located on transmembrane domain helix (TM) 4 and 5. Furthermore, our findings highlight the crucial role of transmembrane domain helix 3 in sensing agonist moieties, representing the pivotal mechanism of receptor activation for both CysLT1R and CysLT2R. Collectively, the insights derived from our structural investigation establish a foundation for comprehending CysLT2R activation by its endogenous ligand LTD4, offering a rational basis for the design of drugs targeting CysLT2R.
Collapse
Affiliation(s)
- Mengting Jiang
- Lingang Laboratory, Shanghai200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - Youwei Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Xiaodong Luan
- Department of Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing100730, China
- Center for Drug Research and Evaluation, National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing100730, China
| | - Kai Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
| | - Zhen Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing210023, China
| | - H. Eric Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
| | - Shuyang Zhang
- Department of Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing100730, China
- Center for Drug Research and Evaluation, National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Beijing100730, China
| | - Yi Jiang
- Lingang Laboratory, Shanghai200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai201210, China
| | - Wanchao Yin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai201203, China
- University of Chinese Academy of Sciences, Beijing100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong528400, China
| |
Collapse
|
2
|
Prakash P, Randolph CE, Walker KA, Chopra G. Lipids: Emerging Players of Microglial Biology. Glia 2025; 73:657-677. [PMID: 39688320 PMCID: PMC11784843 DOI: 10.1002/glia.24654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024]
Abstract
Lipids are small molecule immunomodulators that play critical roles in maintaining cellular health and function. Microglia, the resident immune cells of the central nervous system, regulate lipid metabolism both in the extracellular environment and within intracellular compartments through various mechanisms. For instance, glycerophospholipids and fatty acids interact with protein receptors on the microglial surface, such as the Triggering Receptor Expressed on Myeloid Cells 2, influencing cellular functions like phagocytosis and migration. Moreover, cholesterol is essential not only for microglial survival but, along with other lipids such as fatty acids, is crucial for the formation, function, and accumulation of lipid droplets, which modulate microglial activity in inflammatory diseases. Other lipids, including acylcarnitines and ceramides, participate in various signaling pathways within microglia. Despite the complexity of the microglial lipidome, only a few studies have investigated the effects of specific lipid classes on microglial biology. In this review, we focus on major lipid classes and their roles in modulating microglial function. We also discuss novel analytical techniques for characterizing the microglial lipidome and highlight gaps in current knowledge, suggesting new directions for future research on microglial lipid biology.
Collapse
Affiliation(s)
- Priya Prakash
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Neuroscience Institute, NYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | | | - Gaurav Chopra
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Integrative Neuroscience, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Drug Discovery, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue UniversityWest LafayetteIndianaUSA
- Regenstrief Center for Healthcare Engineering, Purdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
3
|
Ye T, Zhang N, He Y, Chen C, Zha Q, Zhang A, Sun X, Wu X. Electroacupuncture pretreatment inhibits the TLR4/NF-κB/TXNIP/NLRP3 signaling pathway and modulates microglial polarization to alleviate cerebral ischemia-reperfusion injury in rats. Neuroscience 2024; 562:33-42. [PMID: 39424263 DOI: 10.1016/j.neuroscience.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 10/21/2024]
Abstract
Cerebral ischemia-reperfusion injury is frequently associated with neuroinflammation. The modulation of microglial polarization presents a promising approach for addressing cerebral ischemia-reperfusion injury. While electroacupuncture preconditioning has demonstrated efficacy in the management of ischemic stroke, the underlying therapeutic mechanisms remain inadequately understood. The investigation focused on elucidating the relationship between alterations in the TLR4/NF-κB/TXNIP/NLRP3 signaling pathway and microglial polarization subsequent to EA pretreatment. Established a middle cerebral artery occlusion (MCAO) rat model following electroacupuncture (EA) treatment at the Baihui (GV 20) acupoint. Male Sprague-Dawley rats were randomly assigned to the sham, Ischemia/Reperfusion (I/R), I/R + EA groups (n = 6). The results of Nissl Staining and TUNEL Stainingl showed that the number of curative neurons increased significantly after pretreatment, indicating an improvement in neuron formation and an increase in the number of austenite. The level of apoptosis in brain tissue in the I/R group was significantly higher than that in the sham operation group. Electroacupuncture pretreatment can effectively inhibit apoptosis occurrence. In addition, electric acupuncture pretreatment protects rat blood-brain barrier integrity and mitochondrial function. After treatment, the number of M1-type microglia decreased, while the number of M2-type microglia increased. These results suggest that EA preconditioning may alleviate neurological deficits and neuronal apoptosis caused by cerebral I/R injury, while maintaining the integrity of the blood-brain barrier and promoting microglial polarization through the TLR4/NF-κB/TXNIP/NLRP3 signaling pathway. Our findings establish a new molecular mechanism and theoretical foundation for electroacupuncture therapy of ischemic stroke.
Collapse
Affiliation(s)
- Tao Ye
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Ning Zhang
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Yunting He
- The First Clinical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Chunyan Chen
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Qiqi Zha
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Anbang Zhang
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Xiuqi Sun
- Department of Neurology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Xuemei Wu
- Department of Rehabilitation, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China.
| |
Collapse
|
4
|
Zheng X, Hu F, Chen X, Yang G, Li M, Peng Y, Li J, Yang S, Zhang L, Wan J, Wei N, Li R. Role of microglia polarization induced by glucose metabolism disorder in the cognitive impairment of mice from PM 2.5 exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176603. [PMID: 39349199 DOI: 10.1016/j.scitotenv.2024.176603] [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: 06/28/2024] [Revised: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
Studies have found that PM2.5 can damage the brain, accelerate cognitive impairment, and increase the risk of developing a variety of neurodegenerative diseases. However, the potential molecular mechanisms by which PM2.5 causes learning and memory problems are yet to be explored. In this study, we evaluated the neurotoxic effects in mice after 12 weeks of PM2.5 exposure, and found that this exposure resulted in learning and memory disorders, pathological brain damage, and M1 phenotype polarization on microglia, especially in the hippocampus. The severity of this damage increased with increasing PM2.5 concentration. Proteomic analysis, as well as validation results, suggested that PM2.5 exposure led to abnormal glucose metabolism in the mouse brain, which is mainly characterized by significant expression of hexokinase, phosphofructokinase, and lactate dehydrogenase. We therefore administered the glycolysis inhibitor 2-deoxy-d-glucose (2-DG) to the mice exposed to PM2.5, and showed that inhibition of glycolysis by 2-DG significantly alleviated PM2.5-induced hippocampal microglia M1 phenotype polarization, and reduced the release of inflammatory factors, improved synaptic structure and related protein expression, which alleviated the cognitive impairment induced by PM2.5 exposure. In summary, our study found that abnormal glucose metabolism-mediated inflammatory polarization of microglia played a role in learning and memory disorders in mice exposed to PM2.5. This study provides new insights into the neurotoxicity caused by PM2.5 exposure, and provides some theoretical references for the prevention and control of cognitive impairment induced by PM2.5 exposure.
Collapse
Affiliation(s)
- Xinyue Zheng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Fei Hu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xinyue Chen
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Ge Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Min Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Yang Peng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Jinghan Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Shuiqing Yang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Ling Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Jian Wan
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Nianpeng Wei
- Wuhan Hongpeng Ecological Technology Co., Ltd., Wuhan 430070, China
| | - Rui Li
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
5
|
Ma X, Sun Y, Li C, Wang M, Zang Q, Zhang X, Wang F, Niu Y, Hua J. Novel Insights Into DLAT's Role in Alzheimer's Disease-Related Copper Toxicity Through Microglial Exosome Dynamics. CNS Neurosci Ther 2024; 30:e70064. [PMID: 39428563 PMCID: PMC11491298 DOI: 10.1111/cns.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/10/2024] [Accepted: 09/03/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder, with recent research emphasizing the roles of microglia and their secreted extracellular vesicles in AD pathology. However, the involvement of specific molecular pathways contributing to neuronal death in the context of copper toxicity remains largely unexplored. OBJECTIVE This study investigates the interaction between pyruvate kinase M2 (PKM2) and dihydrolipoamide S-acetyltransferase (DLAT), particularly focusing on copper-induced neuronal death in Alzheimer's disease. METHODS Gene expression datasets were analyzed to identify key factors involved in AD-related copper toxicity. The role of DLAT was validated using 5xFAD transgenic mice, while in vitro experiments were conducted to assess the impact of microglial exosomes on neuronal PKM2 transfer and DLAT expression. The effects of inhibiting the PKM2 transfer via microglial exosomes on DLAT expression and copper-induced neuronal death were also evaluated. RESULTS DLAT was identified as a critical factor in the pathology of AD, particularly in copper toxicity. In 5xFAD mice, increased DLAT expression was linked to hippocampal damage and cognitive decline. In vitro, microglial exosomes were shown to facilitate the transfer of PKM2 to neurons, leading to upregulation of DLAT expression and increased copper-induced neuronal death. Inhibition of PKM2 transfer via exosomes resulted in a significant reduction in DLAT expression, mitigating neuronal death and slowing AD progression. CONCLUSION This study uncovers a novel pathway involving microglial exosomes and the PKM2-DLAT interaction in copper-induced neuronal death, providing potential therapeutic targets for Alzheimer's disease. Blocking PKM2 transfer could offer new strategies for reducing neuronal damage and slowing disease progression in AD.
Collapse
Affiliation(s)
- Xiang Ma
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yusheng Sun
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Changchun Li
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Man Wang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Qijiao Zang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Xuxia Zhang
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Feng Wang
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Yulan Niu
- Department of Chemistry and Chemical EngineeringTaiyuan Institute of TechnologyTaiyuanP. R. China
| | - Jiai Hua
- Laboratory of Biochemistry and PharmacyTaiyuan Institute of TechnologyTaiyuanP. R. China
| |
Collapse
|
6
|
Huang SC, Huang HC, Liao WL, Kao ST, Cheng CY. Neuroprotective effects of Gastrodia elata Blume on promoting M2 microglial polarization by inhibiting JNK/TLR4/T3JAM/NF-κB signaling after transient ischemic stroke in rats. Front Pharmacol 2024; 15:1469602. [PMID: 39391701 PMCID: PMC11465390 DOI: 10.3389/fphar.2024.1469602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Background Gastrodia elata Blume, also called Tian Ma (TM), has been used to treat stroke for centuries. However, its effects on inflammation in acute cerebral ischemic injury and underlying mechanisms involved in microglial polarization remain unknown. The present study explored the effects of the TM extract on the modulation of microglial M1/M2 polarization 2 days after transient cerebral ischemia. Methods Male Sprague Dawley rats were intracerebroventricularly administered with 1% dimethyl sulfoxide 25 min before cerebral ischemia and subsequently intraperitoneally administered 0.25 g/kg (DO + TM-0.25 g), 0.5 g/kg (DO + TM-0.5 g), or 1 g/kg (DO + TM-1 g) of the TM extract after cerebral ischemia onset. Results DO + TM-0.5 g and DO + TM-1 g treatments downregulated the following: phospho-c-Jun N-terminal kinase (p-JNK)/JNK, tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3), TRAF3-interacting JNK-activating modulator (T3JAM), p-nuclear factor-kappa B p65 (p-NF-κB p65)/NF-κB p65, ionized calcium-binding adapter molecule 1 (Iba1), CD86, TNF-α, interleukin (IL)-1β, and IL-6 expression and toll-like receptor 4 (TLR4)/Iba1, CD86/Iba1, and p-NF-κB p65/Iba1 coexpression. These treatments also upregulated IL-10, nerve growth factor, and vascular endothelial growth factor A expression and YM-1/2/Iba1 and IL-10/neuronal nuclei coexpression in the cortical ischemic rim. The JNK inhibitor SP600125 exerted similar treatment effects as the DO + TM-0.5 g and DO + TM-1 g treatments. Conclusion DO + TM-0.5 g and DO + TM-1 g/kg treatments attenuate cerebral infarction by inhibiting JNK-mediated signaling. TM likely exerts the neuroprotective effects of promoting M1 to M2 microglial polarization by inhibiting JNK/TLR4/T3JAM/NF-κB-mediated signaling in the cortical ischemic rim 2 days after transient cerebral ischemia.
Collapse
Affiliation(s)
- Shang-Chih Huang
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Hui-Chi Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chin-Yi Cheng
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung, Taiwan
| |
Collapse
|
7
|
Cui Y, Cui M, Wang L, Wang N, Chen Y, Lv S, Zhang L, Chen C, Yang Y, Wang F, Wang L, Cui H. Huanglian Jiedu decoction alleviates ischemia-induced cerebral injury in rats by mitigating NET formation and activiting GABAergic synapses. J Cell Mol Med 2024; 28:e18528. [PMID: 39099086 PMCID: PMC11298410 DOI: 10.1111/jcmm.18528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 08/06/2024] Open
Abstract
Huanglian Jiedu decoction (HLJD) has been used to treat ischemic stroke in clinic. However, the detailed protective mechanisms of HLJD on ischemic stroke have yet to be elucidated. The aim of this study is to elucidate the underlying pharmacological mechanisms of HLJD based on the inhibition of neuroinflammation and the amelioration of nerve cell damage. A middle cerebral artery occlusion reperfusion (MCAO/R) model was established in rats and received HLJD treatment. Effects of HLJD on neurological function was assessed based on Bederson's score, postural reflex test and asymmetry score. 2, 3, 5-Triphenyltetrazolium chloride (TTC) staining, Hematein and eosin (HE) and Nissl staining were used to observe the pathological changes in brain. Then, transcriptomics was used to screen the differential genes in brain tissue in MCAO/R model rats following HLJD intervention. Subsequently, the effects of HLJD on neutrophil extracellular trap (NET) formation-related neuroinflammation, gamma-aminobutyric acid (GABA)ergic synapse activation, nerve cell damage and proliferation were validated using immunofluorescence, western blot and enzyme-linked immunosorbent assay (ELISA). Our results showed that HLJD intervention reduced the Bederson's score, postural reflex test score and asymmetry score in MCAO/R model rats. Pathological staining indicated that HLJD treatment decreased the cerebral infarction area, mitigated neuronal damage and increased the numbers of Nissl bodies. Transcriptomics suggested that HLJD affected 435 genes in MCAO/R rats. Among them, several genes involving in NET formation and GABAergic synapses pathways were dysregulated. Subsequent experimental validation showed that HLJD reduced the MPO+CitH3+ positive expression area, reduced the protein expression of PAD4, p-P38/P38, p-ERK/ERK and decreased the levels of IL-1β, IL-6 and TNF-α, reversed the increase of Iba1+TLR4+, Iba1+p65+ and Iba1+NLRP3+ positive expression area in brain. Moreover, HLJD increased GABA levels, elevated the protein expression of GABRG1 and GAT3, decreased the TUNEL positive expression area and increased the Ki67 positive expression area in brain. HLJD intervention exerts a multifaceted positive impact on ischemia-induced cerebral injury in MCAO/R rats. This intervention effectively inhibits neuroinflammation by mitigating NET formation, and concurrently improves nerve cell damage and fosters nerve cell proliferation through activating GABAergic synapses.
Collapse
Affiliation(s)
- Youxiang Cui
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Mingyue Cui
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Leilei Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Ning Wang
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Yao Chen
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| | - Shuquan Lv
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Limin Zhang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Congai Chen
- Beijing University of Chinese MedicineBeijingChina
| | - Yanwen Yang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Feng Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Lichun Wang
- Key Laboratory of Neurological RehabilitationCangzhou Hospital of Integrated Traditional Chinese Medicine and Western MedicineCangzhouChina
| | - Huantian Cui
- First School of Clinical MedicineYunnan University of Chinese MedicineKunmingChina
| |
Collapse
|
8
|
Anwar MM, Pérez-Martínez L, Pedraza-Alva G. Exploring the Significance of Microglial Phenotypes and Morphological Diversity in Neuroinflammation and Neurodegenerative Diseases: From Mechanisms to Potential Therapeutic Targets. Immunol Invest 2024; 53:891-946. [PMID: 38836373 DOI: 10.1080/08820139.2024.2358446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Studying various microglial phenotypes and their functions in neurodegenerative diseases is crucial due to the intricate nature of their phenomics and their vital immunological role. Microglia undergo substantial phenomic changes, encompassing morphological, transcriptional, and functional aspects, resulting in distinct cell types with diverse structures, functions, properties, and implications. The traditional classification of microglia as ramified, M1 (proinflammatory), or M2 (anti-inflammatory) phenotypes is overly simplistic, failing to capture the wide range of recently identified microglial phenotypes in various brain regions affected by neurodegenerative diseases. Altered and activated microglial phenotypes deviating from the typical ramified structure are significant features of many neurodegenerative conditions. Understanding the precise role of each microglial phenotype is intricate and sometimes contradictory. This review specifically focuses on elucidating recent modifications in microglial phenotypes within neurodegenerative diseases. Recognizing the heterogeneity of microglial phenotypes in diseased states can unveil novel therapeutic strategies for targeting microglia in neurodegenerative diseases. Moreover, the exploration of the use of healthy isolated microglia to mitigate disease progression has provided an innovative perspective. In conclusion, this review discusses the dynamic landscape of mysterious microglial phenotypes, emphasizing the need for a nuanced understanding to pave the way for innovative therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai M Anwar
- Department of Biochemistry, National Organization for Drug Control and Research (NODCAR)/Egyptian Drug Authority (EDA), Cairo, Egypt
| | - Leonor Pérez-Martínez
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| | - Gustavo Pedraza-Alva
- Neuroimmunobiology Laboratory, Department of Molecular Medicine and Bioprocesses, Institute of Biotechnology, National Autonomous University of Mexico, Cuernavaca, Morelos, Mexico
| |
Collapse
|
9
|
Sahebi K, Foroozand H, Amirsoleymani M, Eslamzadeh S, Negahdaripour M, Tajbakhsh A, Rahimi Jaberi A, Savardashtaki A. Advancing stroke recovery: unlocking the potential of cellular dynamics in stroke recovery. Cell Death Discov 2024; 10:321. [PMID: 38992073 PMCID: PMC11239950 DOI: 10.1038/s41420-024-02049-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Stroke stands as a predominant cause of mortality and morbidity worldwide, and there is a pressing need for effective therapies to improve outcomes and enhance the quality of life for stroke survivors. In this line, effective efferocytosis, the clearance of apoptotic cells, plays a crucial role in neuroprotection and immunoregulation. This process involves specialized phagocytes known as "professional phagocytes" and consists of four steps: "Find-Me," "Eat-Me," engulfment/digestion, and anti-inflammatory responses. Impaired efferocytosis can lead to secondary necrosis and inflammation, resulting in adverse outcomes following brain pathologies. Enhancing efferocytosis presents a potential avenue for improving post-stroke recovery. Several therapeutic targets have been identified, including osteopontin, cysteinyl leukotriene 2 receptor, the µ opioid receptor antagonist β-funaltrexamine, and PPARγ and RXR agonists. Ferroptosis, defined as iron-dependent cell death, is now emerging as a novel target to attenuate post-stroke tissue damage and neuronal loss. Additionally, several biomarkers, most importantly CD163, may serve as potential biomarkers and therapeutic targets for acute ischemic stroke, aiding in stroke diagnosis and prognosis. Non-pharmacological approaches involve physical rehabilitation, hypoxia, and hypothermia. Mitochondrial dysfunction is now recognized as a major contributor to the poor outcomes of brain stroke, and medications targeting mitochondria may exhibit beneficial effects. These strategies aim to polarize efferocytes toward an anti-inflammatory phenotype, limit the ingestion of distressed but viable neurons, and stimulate efferocytosis in the late phase of stroke to enhance post-stroke recovery. These findings highlight promising directions for future research and development of effective stroke recovery therapies.
Collapse
Affiliation(s)
- Keivan Sahebi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hassan Foroozand
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Saghi Eslamzadeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manica Negahdaripour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Abbas Rahimi Jaberi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Liu C, Yin T, Zhang M, Li Z, Xu B, Lv H, Wang P, Wang J, Hao J, Zhang L. Function of miR-21-5p derived from ADSCs-exos on the neuroinflammation after cerebral ischemia. J Stroke Cerebrovasc Dis 2024; 33:107779. [PMID: 38768666 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107779] [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: 10/12/2023] [Revised: 02/13/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024] Open
Abstract
INTRODUCTION Cerebral ischemia (CI) induces a profound neuroinflammatory response, but the underlying molecular mechanism remains unclear. Exosomes from adipose-derived stem cells (ADSC-exos) have been found to play a crucial role in cell communication by transferring molecules including microRNAs (miRNAs), which have been shown to modulate the inflammatory response after CI and are viable molecular targets for altering brain function. The current study aimed to explore the contribution of ADSC-exosomal miR-21-5p to the neuroinflammation after CI. METHODS The differentially expressed miR-21-5p in CI was screened based on literature search. The target mRNAs of miR-21-5p were predicted using online databases and verified by luciferase reporter assay. Then, BV2 cells were treated with hemin to simulate the inflammatory response after CI, and its animal model was induced using the MCAO method. Ischemia was evaluated in rats using 2, 3, 5-triphenyl tetrazolium chloride (TTC) staining. ADSCs-exos were further isolated and identified by western blot analysis and transmission electron microscope. RESULTS MiR-21-5p was significantly down-regulated in CI and alleviated neuropathic damage after CI by the PIK3R1/PI3K/AKT signaling axis. And miR-21-5p derived from ADSCs-exos alleviated neuroinflammation after CI via promoting microglial M2 polarization. CONCLUSION We demonstrated that ADSC-exosomal miR-21-5p mitigated post-CI inflammatory response through the PIK3R1/PI3K/AKT signaling axis and could offer neuroprotection after CI through promoting polarization of M2 microglia.
Collapse
Affiliation(s)
- Chao Liu
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Tengkun Yin
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Meng Zhang
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Zhongchen Li
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Bin Xu
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Hang Lv
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Peijian Wang
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Jiheng Hao
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng Brain Hospital, No. 45 Huashan Road, Dongchangfu District, Liaocheng, Shandong 252000, China.
| |
Collapse
|
11
|
Lu W, Wang Y, Wen J. The Roles of RhoA/ROCK/NF-κB Pathway in Microglia Polarization Following Ischemic Stroke. J Neuroimmune Pharmacol 2024; 19:19. [PMID: 38753217 DOI: 10.1007/s11481-024-10118-w] [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: 11/18/2023] [Accepted: 04/21/2024] [Indexed: 05/21/2024]
Abstract
Ischemic stroke is the leading cause of death and disability worldwide. Nevertheless, there still lacks the effective therapies for ischemic stroke. Microglia are resident macrophages of the central nervous system (CNS) and can initiate immune responses and monitor the microenvironment. Microglia are activated and polarize into proinflammatory or anti‑inflammatory phenotype in response to various brain injuries, including ischemic stroke. Proinflammatory microglia could generate immunomodulatory mediators, containing cytokines and chemokines, these mediators are closely associated with secondary brain damage following ischemic stroke. On the contrary, anti-inflammatory microglia facilitate recovery following stroke. Regulating the activation and the function of microglia is crucial in exploring the novel treatments for ischemic stroke patients. Accumulating studies have revealed that RhoA/ROCK pathway and NF-κB are famous modulators in the process of microglia activation and polarization. Inhibiting these key modulators can promote the polarization of microglia to anti-inflammatory phenotype. In this review, we aimed to provide a comprehensive overview on the role of RhoA/ROCK pathway and NF-κB in the microglia activation and polarization, reveal the relationship between RhoA/ROCK pathway and NF-κB in the pathological process of ischemic stroke. In addition, we likewise discussed the drug modulators targeting microglia polarization.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Medical Branch, Hefei Technology College, Hefei, China
| | - Yilin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
Yu Y, Liao X, Xie X, Li Q, Chen X, Liu R. The role of neuroglial cells communication in ischemic stroke. Brain Res Bull 2024; 209:110910. [PMID: 38423190 DOI: 10.1016/j.brainresbull.2024.110910] [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: 11/10/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Ischemic stroke is one of the leading causes of death and disability globally, but its treatment options are limited due to therapeutic window and reperfusion injury constraints. Microglia, astrocytes, and oligodendrocytes are the major components of the neurovascular unit, and there is substantial evidence suggesting their contributions to maintaining homeostasis in the central nervous system. Neuroglial cells participate in neuronal physiological functions and the repair of damaged neurons through various communication methods, including gap junctions, chemical signaling, and extracellular vesicles, in conjunction with other components of the neurovascular unit. Ischemia-induced microglia and astrocytes polarize into "M1/M2" and "A1/A2" phenotypes and exert neurotoxic or neuroprotective effects by releasing soluble factors, secreting extracellular vesicles, and forming syncytia networks in the acute (<72 h), subacute (>72 h), and chronic phases (>6 weeks). Apoptosis of oligodendrocytes due to ischemic hypoxia leads to white matter injury, causing long-term cognitive dysfunction, and promoting oligodendrogenesis is a crucial direction for achieving functional recovery in ischemic stroke. In this article, we summarize the cellular interactions following cerebral ischemia, analyze the roles of neuroglial cells through gap junctions, chemical signaling, and extracellular vesicles in different stages of ischemic stroke, and further explore strategies for intervening in ischemic stroke.
Collapse
Affiliation(s)
- Yunling Yu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xinglan Liao
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xinyu Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Qihua Li
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Xuehong Chen
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Ruizhen Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
13
|
Augusto-Oliveira M, Tremblay MÈ, Verkhratsky A. Receptors on Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:83-121. [PMID: 39207688 DOI: 10.1007/978-3-031-55529-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglial cells are the most receptive cells in the central nervous system (CNS), expressing several classes of receptors reflecting their immune heritage and newly acquired neural specialisation. Microglia possess, depending on the particular context, receptors to neurotransmitters and neuromodulators as well as immunocompetent receptors. This rich complement allows microglial cells to monitor the functional status of the nervous system, contribute actively to the regulation of neural activity and plasticity and homeostasis, and guard against pathogens as well as other challenges to the CNS's integrity and function.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
14
|
Shi J, Wang X, Kang C, Liu J, Ma C, Yang L, Hu J, Zhao N. TREM2 regulates BV2 microglia activation and influences corticosterone-induced neuroinflammation in depressive disorders. Brain Res 2024; 1822:148664. [PMID: 37923002 DOI: 10.1016/j.brainres.2023.148664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/14/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Depressive disorders is a serious mental illness, and its underlying pathological mechanisms remain unclear. The overactivation of microglia and neuroinflammation are thought to play an essential role in the occurrence and development of depressive disorders. TREM2, an immune protein mainly expressed in microglia, is an important part of nerve cells involved in inflammatory response. Corticosterone (CORT) is often referred to as a stress hormone and plays a role in the immune system and stress response. Therefore, this study investigated the role of TREM2 in CORT-induced BV2 cell damage and preliminarily analyzed the effects of TREM2 on JAK2/STAT3 signaling pathway and microglia polarization. The cell model of CORT-induced depression in vitro was established, and the effect of CORT on the activity of BV2 microglia was detected by CCK8. Plasmid transfection was used to overexpress and interfere with TREM2 in BV2 cells cultured by CORT. Western blotting, PCR, and ELISA analyzed the expression of related proteins and inflammatory factors. The results showed that CORT could affect BV2 cell proliferation and TREM2 levels. In the presence of CORT, overexpression of TREM2 decreased the levels of TNF-α, IL-1β, and IL-6 and increased the levels of IL-10. Interference with TREM2 increased the levels of TNF-α, IL-1β, and IL-6 and decreased the levels of IL-10. TREM2 can affect the release of inflammatory factors through the JAK2/STAT3 signaling pathway and regulate the M1/M2 phenotypic transformation of microglia. TREM2 plays a role in regulating CORT-induced inflammatory responses, revealing the influence of TREM2 on the neuroinflammatory pathogenesis of depressive disorders and suggesting that TREM2 may be a new target for the prevention and treatment of depressive disorders.
Collapse
Affiliation(s)
- Jingjing Shi
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Xiaohong Wang
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Chuanyi Kang
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Jiacheng Liu
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Caina Ma
- Harbin First Specialized Hospital, Heilongjiang Province, China
| | - Liying Yang
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China
| | - Jian Hu
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China.
| | - Na Zhao
- Department of Psychiatry, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
15
|
Shin MJ, Lee CS, Kim SH. Screening for Lactic Acid Bacterial Strains as Probiotics Exhibiting Anti-inflammatory and Antioxidative Characteristic Via Immune Modulation in HaCaT Cell. Probiotics Antimicrob Proteins 2023; 15:1665-1680. [PMID: 36806154 DOI: 10.1007/s12602-023-10048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
In this study, the basic probiotic characteristics and functional properties of lactic acid bacteria (LAB) were investigated using two in vitro models of inflammation induced by lipopolysaccharide (LPS) and H2O2. Fifteen strains were prescreened out of 60 LAB candidates based on their radical scavenging activity to determine the antioxidant capacity of the strains. The top 15 candidates were further investigated to evaluate their survival rate under low pH and bile salt conditions that mimic the intestinal environment. Three strains, Levilactobacillus brevis D70 (Levilact), Lactiplantibacillus pentosus S16 (Lactipla), and Limosilactobacillus fermentum MF10 (Limosilact), were capable of scavenging free radicals and survived under artificial intestinal conditions. Therefore, Levilact. brevis D70, Lactipla. pentosus S16, and Limosilact. fermentum MF10 were selected for further antioxidant, anti-inflammation, and mitochondrial activity examinations via cell models of inflammation and oxidative stress. Among the three strains, Limosilact. fermentum MF10 showed the highest anti-inflammatory activities by significantly downregulating the relative mRNA expression levels of inflammatory biomarkers such as interleukin 8 (IL-8) and interferon-gamma (IFN-γ) induced by LPS (P < 0.05). Moreover, Limosilact. fermentum MF10 was also capable of upregulating the gene expression levels of antioxidative mediator glutathione peroxidase 4 (GPX4) induced by reactive oxygen species (ROS) in both human HT-29 epithelial cells and human HaCaT keratinocytes. Limosilact. fermentum MF10 was also capable of regulating mitochondrial membrane potential (MMP), which plays a key role in the mitochondrial activity of HaCaT cells. As a result, Limosilact. fermentum MF10 showed the highest potential for probiotic properties and impacts the immune-related gut-skin axis by altering proinflammatory cytokines, antioxidative biomarkers, and MMP.
Collapse
Affiliation(s)
- Min Jae Shin
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Chul Sang Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Life Science and Natural Resources, Korea University, Seoul, 02841, Republic of Korea.
| | - Sae Hun Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Life Science and Natural Resources, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
16
|
Li Y, Li YJ, Zhu ZQ. To re-examine the intersection of microglial activation and neuroinflammation in neurodegenerative diseases from the perspective of pyroptosis. Front Aging Neurosci 2023; 15:1284214. [PMID: 38020781 PMCID: PMC10665880 DOI: 10.3389/fnagi.2023.1284214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and motor neuron disease, are diseases characterized by neuronal damage and dysfunction. NDs are considered to be a multifactorial disease with diverse etiologies (immune, inflammatory, aging, genetic, etc.) and complex pathophysiological processes. Previous studies have found that neuroinflammation and typical microglial activation are important mechanisms of NDs, leading to neurological dysfunction and disease progression. Pyroptosis is a new mode involved in this process. As a form of programmed cell death, pyroptosis is characterized by the expansion of cells until the cell membrane bursts, resulting in the release of cell contents that activates a strong inflammatory response that promotes NDs by accelerating neuronal dysfunction and abnormal microglial activation. In this case, abnormally activated microglia release various pro-inflammatory factors, leading to the occurrence of neuroinflammation and exacerbating both microglial and neuronal pyroptosis, thus forming a vicious cycle. The recognition of the association between pyroptosis and microglia activation, as well as neuroinflammation, is of significant importance in understanding the pathogenesis of NDs and providing new targets and strategies for their prevention and treatment.
Collapse
Affiliation(s)
- Yuan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- College of Anesthesiology, Zunyi Medical University, Zunyi, China
| | - Ying-Jie Li
- Department of General Surgery, Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| | - Zhao-Qiong Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
17
|
Zhang XH, Cui H, Zheng SM, Lu Y, Yuan HW, Zhang L, Wang HH, Du RS. Electroacupuncture regulates microglial polarization via inhibiting NF-κB/COX2 pathway following traumatic brain injury. Brain Res 2023; 1818:148516. [PMID: 37562566 DOI: 10.1016/j.brainres.2023.148516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Neuroinflammation and oxidative stress are important pathological mechanisms following traumatic brain injury (TBI). The NF-κB/COX2 pathway regulates neuroinflammation and oxidative damage, while microglia also play an important role in neuroinflammation. Since NF-κB is involved in microglial polarization, targeting this pathway and microglial polarization is a critical component of TBI treatment. Currently, electroacupuncture (EA) is widely used to treat various symptoms after TBI, but the mechanisms of EA remain poorly understood. Additionally, the optimal frequency of EA remains unclear, which affects its efficacy. This study focuses on exploring the optimal frequency parameters of EA on TBI and investigating the underlying mechanisms of EA through NF-κB/COX2 pathway and microglial polarization. METHODS The study was divided into two parts. In Experiment 1, 42 Sprague Dawley (SD) rats were induced and randomly divided into seven groups (n = 6). Except for the sham group, all rats underwent controlled cortical impact (CCI) to establish TBI model. Four EA groups (with different frequencies) and manual acupuncture (without current stimulation) received stimulation on the acupoints of Shuigou (GV26), Fengchi (GB20) and Neiguan (PC6) once a day for 7 days. The neurological function was assessed by modified Neurological Severity Scores (mNSS), and the rats' memory and learning were examined by the Morris water maze (MWM). SOD, MDA, and GSH-Px were detected to evaluate the levels of oxidative stress. The levels of IL-1β, IL-6, and TNF-α were evaluated by Enzyme Linked Immunosorbent Assay (ELISA). Detection of the above indicators indicated a treatment group that exerted the strongest neuroprotection against TBI, we then conducted Experiment 2 using this screened acupuncture treatment to investigate the mechanism of acupuncture. 48 rats were randomly divided into four groups (n = 12): sham, TBI model, acupuncture and PDTC (NF-κB inhibitor). Evaluations of mNSS, MWM test, SOD, MDA, GSH-Px, IL-1β, IL-6, TNF-α, and IL-10 were the same as in Experiment 1. Western blot was applied for detecting the expression levels of NF-κB, p-NF-κB, COX2, and Arg-1. TUNEL was used to examine neuronal apoptosis. Brain structure was observed by H&E. Iba-1, COX2, and Arg-1 were investigated by immunofluorescence staining. RESULTS EA with frequency of 2/100 Hz markedly improved neuronal and cognitive function as compared to the other treatment groups. Moreover, it downregulated the expression of MDA, IL-6, IL-1β, and TNF-α and upregulated the levels of SOD and GSH-Px. In addition, Both EA with 2/100 Hz and PDTC reduced the levels of p-NF-κB, COX2 and M1 markers (COX2, IL-6, IL-1β, TNF-α) and increased the levels of M2 markers (Arg-1, IL-10). Moreover, they had similar effects on reducing inflammation, oxidative stress and apoptosis, and improving neuronal and cognitive function. CONCLUSIONS The collective findings strongly suggest that EA with 2/100 Hz can improve neurologic function by suppressing neuroinflammation, oxidative stress and apoptosis. Additionally, we confirm that EA promotes microglial polarization towards the M2 phenotype through the suppression of NF-κB/COX2 pathway, thus exerting neuroprotective effects after TBI.
Collapse
Affiliation(s)
- Xiao-Hui Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Hai Cui
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Shu-Mei Zheng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Hong-Wen Yuan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Lu Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Hong-Hong Wang
- Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, China
| | - Ruo-Sang Du
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
18
|
Wang X, Shi YJ, Niu TY, Chen TT, Li HB, Wu SH, Li GL. Neuroprotective effect of 20 (S) - Protopanaxadiol (PPD) attenuates NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia rats. Neurosci Lett 2023; 814:137439. [PMID: 37579868 DOI: 10.1016/j.neulet.2023.137439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
20(S)-protopanaxadiol (PPD), one of the ginsenosides from Panax ginseng, has been reported to improve performance with dementia. This study aimed to investigate the neuroprotective effect of PPD attenuating NLRP3 inflammasome-mediated microglial pyroptosis in vascular dementia (VD) rats induced by bilateral common carotid artery ligation (2-VO). Male Sprague-Dawley rats (SPF, 150-180 g, n = 10/group) were randomly divided into PPD (20, 10, 5 mg/kg, subcutaneous injection once per day for 3 weeks), model, and vehicle-sham group. It was found that PPD significantly reversed 2-VO-induced cognitive impairment by decreasing escape latency and spontaneous alternation and increasing the number of crossing platforms, showing memory-improving effects. PPD improved the pathological morphology of brain tissue in VD rats. PPD significantly reduced the cerebral infarction area and the activation of microglia in the cortex and hippocampal DG, CA1, and CA3 area. Moreover, PPD could attenuate NLRP3 inflammasome-mediated microglial pyroptosis, inhibit the positive expression of NLRP3, decrease IL-1β, and IL-18 levels, and increase IL-10 levels in the brain cortex. PPD also significantly alleviated the neurotoxicity by decreasing the Aβ and p-Tau in hippocampal DG, CA1, and CA3 areas. In addition, the levels of NLRP3, ASC, and IL-1β in the cortex, APP, BACE1, and p-Tau in the hippocampus were significantly reduced by PPD. These results suggested that PPD hinders microglial activation to alleviate neuroinflammation of NLRP3 inflammasome and inhibits neurotoxicity of Aβ deposition and Tau phosphorylation in 2-VO-induced VD rats.
Collapse
Affiliation(s)
- Xue Wang
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ya-Jin Shi
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ting-Yuan Niu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Ting-Ting Chen
- Pharmacy College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Han-Bing Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China
| | - Su-Hui Wu
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| | - Gen-Lin Li
- Medical College, Henan University of Chinese Medicine, Zheng-Zhou 450046, China.
| |
Collapse
|
19
|
Dai M, Zhang Y, Jiao Y, Deng Y, Du X, Yang C. Immunomodulatory effects of one novel microRNA miR-63 in pearl oyster Pinctada fucata martensii. FISH & SHELLFISH IMMUNOLOGY 2023; 140:109002. [PMID: 37586600 DOI: 10.1016/j.fsi.2023.109002] [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: 05/27/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/18/2023]
Abstract
Novel microRNA miR-63 (novel-miR-63) from pearl oyster Pinctada fucata martensii (Pm-novel-miR-63) is a species-specific miRNA. Our previous research has shown that the expression of Pm-novel-miR-63 was significantly downregulated at 24 h after nucleus transplantation. In this study, we analyzed the function and regulatory role of Pm-novel-miR-63 in the immune response of pearl oysters. The results showed that Pm-novel-miR-63 expression increased after the stimulation of pathogen associated molecular patterns at 6-12 h, and the activity of immune and antioxidant enzymes in the serum decreased after Pm-novel-miR-63 overexpression. Transcriptome analysis revealed that Pm-novel-miR-63 participated in regulating transplantation immunity through the Notch and mRNA surveillance signaling pathways. Target prediction and dual luciferase analysis revealed that Pm-GDP-FucTP, Pm-CysLTR2, and Pm-RLR were the target genes of Pm-novel-miR-63. These results suggested that Pm-novel-miR-63 participated in regulating the immune response in pearl oysters and can serve as a new interference target to reasonably control excessive immune rejection in pearl culture.
Collapse
Affiliation(s)
- Meiqi Dai
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Yuting Zhang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Yu Jiao
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China.
| | - Yuewen Deng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524088, China; Guangdong Marine Ecology Early Warning and Monitoring Laboratory, Zhanjiang, 524088, China
| | - Xiaodong Du
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China
| | - Chuangye Yang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524025, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Marine Ecology Early Warning and Monitoring Laboratory, Zhanjiang, 524088, China
| |
Collapse
|
20
|
Chen HC, Chang WC, Chuang JY, Chang KY, Liou JP, Hsu TI. The complex role of eicosanoids in the brain: Implications for brain tumor development and therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:188957. [PMID: 37488051 DOI: 10.1016/j.bbcan.2023.188957] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Eicosanoids are a family of bioactive lipids that play diverse roles in the normal physiology of the brain, including neuronal signaling, synaptic plasticity, and regulation of cerebral blood flow. In the brain, eicosanoids are primarily derived from arachidonic acid, which is released from membrane phospholipids in response to various stimuli. Prostaglandins (PGs) and leukotrienes (LTs) are the major classes of eicosanoids produced in the brain, and they act through specific receptors to modulate various physiological and pathological processes. Dysregulation of eicosanoids has been implicated in the development and progression of brain tumors, including glioblastoma (GBM), meningioma, and medulloblastoma. Eicosanoids have been shown to promote tumor cell proliferation, migration, invasion, angiogenesis, and resistance to therapy. Particularly, PGE2 promotes GBM cell survival and resistance to chemotherapy. Understanding the role of eicosanoids in brain tumors can inform the development of diagnostic and prognostic biomarkers, as well as therapeutic strategies that target eicosanoid pathways. Cyclooxygenase (COX)-2 and 5-lipoxygenase (LOX) inhibitors have been shown to reduce the growth and invasiveness of GBM cells. Moreover, eicosanoids have immunomodulatory effects that can impact the immune response to brain tumors. Understanding the role of eicosanoids in the immune response to brain tumors can inform the development of immunotherapy approaches for these tumors. Overall, the complex role of eicosanoids in the brain underscores the importance of further research to elucidate their functions in normal physiology and disease, and highlights the potential for developing novel therapeutic approaches that target eicosanoid pathways in brain tumors.
Collapse
Affiliation(s)
- Hsien-Chung Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Chang Chang
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Jian-Ying Chuang
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
| | - Jing-Ping Liou
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; TMU Research Center for Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
21
|
Pereira JF, de Sousa Neves JC, Fonteles AA, Bezerra JR, Pires RC, da Silva ATA, Lima FAV, Neves KRT, Oriá RB, de Barros Viana GS, Tavares J, de Sousa Nascimento T, Oliveira AV, Parente ACB, Gomes JMP, de Andrade GM. Palmatine, a natural alkaloid, attenuates memory deficits and neuroinflammation in mice submitted to permanent focal cerebral ischemia. J Neuroimmunol 2023; 381:578131. [PMID: 37413943 DOI: 10.1016/j.jneuroim.2023.578131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/16/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023]
Abstract
Ischemic stroke is one of the major causes of human morbidity and mortality. The pathophysiology of ischemic stroke involves complex events, including oxidative stress and inflammation, that lead to neuronal loss and cognitive deficits. Palmatine (PAL) is a naturally occurring (Coptidis rhizome) isoquinoline alkaloid that belongs to the class of protoberberines and has a wide spectrum of pharmacological and biological effects. In the present study, we evaluated the impact of Palmatine on neuronal damage, memory deficits, and inflammatory response in mice submitted to permanent focal cerebral ischemia induced by middle cerebral artery (pMCAO) occlusion. The animals were treated with Palmatine (0.2, 2 and 20 mg/kg/day, orally) or vehicle (3% Tween + saline solution) 2 h after pMCAO once daily for 3 days. Cerebral ischemia was confirmed by evaluating the infarct area (TTC staining) and neurological deficit score 24 h after pMCAO. Treatment with palmatine (2 and 20 mg/kg) reduced infarct size and neurological deficits and prevented working and aversive memory deficits in ischemic mice. Palmatine, at a dose of 2 mg/kg, had a similar effect of reducing neuroinflammation 24 h after cerebral ischemia, decreasing TNF-, iNOS, COX-2, and NF- κB immunoreactivities and preventing the activation of microglia and astrocytes. Moreover, palmatine (2 mg/kg) reduced COX-2, iNOS, and IL-1β immunoreactivity 96 h after pMCAO. The neuroprotective properties of palmatine make it an excellent adjuvant treatment for strokes due to its inhibition of neuroinflammation.
Collapse
Affiliation(s)
- Juliana Fernandes Pereira
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil
| | - Juliana Catharina de Sousa Neves
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Analu Aragão Fonteles
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Jéssica Rabelo Bezerra
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Rayssa Costa Pires
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Ana Thais Araújo da Silva
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Francisco Arnaldo Viana Lima
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Kelly Rose Tavares Neves
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil
| | - Reinaldo Barreto Oriá
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil
| | - Glauce Socorro de Barros Viana
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Juliete Tavares
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Tyciane de Sousa Nascimento
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Alfaete Vieira Oliveira
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Ana Caroline Barros Parente
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Jessica Maria Pessoa Gomes
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil
| | - Geanne Matos de Andrade
- Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Brazil, Rua Professor Costa Mendes, 1608, 60.430-140 Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Coronel Nunes de Melo, 1127, 60.430-275 Fortaleza, CE, Brazil; Neuroscience and Behavior Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceara, Rua Coronel Nunes de Melo, 1000, 60.430-275 Fortaleza, CE, Brazil.
| |
Collapse
|
22
|
Brunner SM, Schrödl F, Preishuber-Pflügl J, Runge C, Koller A, Lenzhofer M, Reitsamer HA, Trost A. Distribution of the cysteinyl leukotriene system components in the human, rat and mouse eye. Exp Eye Res 2023; 232:109517. [PMID: 37211287 DOI: 10.1016/j.exer.2023.109517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
The cysteinyl leukotrienes (CysLTs) have important functions in the regulation of inflammation and cellular stress. Blocking the CysLT receptors (CysLTRs) with specific antagonists is beneficial against progression of retinopathies (e.g. diabetic retinopathy, wet AMD). However, the exact cellular localization of the CysLTRs and their endogenous ligands in the eye have not been elucidated in detail yet. It is also not known whether the expression patterns differ between humans and animal models. Therefore, the present study aimed to describe and compare the distribution of two important enzymes in CysLT biosynthesis, 5-lipoxygenase (5-LOX) and 5-lipoxygenase-activating protein (FLAP), and of CysLTR1 and CysLTR2 in healthy human, rat and mouse eyes. Human donor eyes (n = 10) and eyes from adult Sprague Dawley rats (n = 5) and CD1 mice (n = 8) of both sexes were collected. The eyes were fixed in 4% paraformaldehyde and cross-sections were investigated by immunofluorescence with specific antibodies against 5-LOX, FLAP (human tissue only), CysLTR1 and CysLTR2. Flat-mounts of the human choroid were prepared and processed similarly. Expression patterns were assessed and semiquantitatively evaluated using a confocal fluorescence microscope (LSM710, Zeiss). We observed so far unreported expression sites for CysLT system components in various ocular tissues. Overall, we detected expression of 5-LOX, CysLTR1 and CysLTR2 in the human, rat and mouse cornea, conjunctiva, iris, lens, ciliary body, retina and choroid. Importantly, expression profiles of CysLTR1 and CysLTR2 were highly similar between human and rodent eyes. FLAP was expressed in all human ocular tissues except the lens. Largely weak immunoreactivity of FLAP and 5-LOX was observed in a few, yet unidentified, cells of diverse ocular tissues, indicating low levels of CysLT biosynthesis in healthy eyes. CysLTR1 was predominantly detected in ocular epithelial cells, supporting the involvement of CysLTR1 in stress and immune responses. CysLTR2 was predominantly expressed in neuronal structures, suggesting neuromodulatory roles of CysLTR2 in the eye and revealing disparate functions of CysLTRs in ocular tissues. Taken together, we provide a comprehensive protein expression atlas of CysLT system components in the human and rodent eye. While the current study is purely descriptive and therefore does not allow significant functional conclusions yet, it represents an important basis for future studies in diseased ocular tissues in which distribution patterns or expression levels of the CysLT system might be altered. Furthermore, this is the first comprehensive study to elucidate expression patterns of CysLT system components in human and animal models that will help to identify and understand functions of the system as well as mechanisms of action of potential CysLTR ligands in the eye.
Collapse
Affiliation(s)
- Susanne M Brunner
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Falk Schrödl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria.
| | - Julia Preishuber-Pflügl
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Christian Runge
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria; Cornea Eye Bank, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Andreas Koller
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Markus Lenzhofer
- Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Herbert A Reitsamer
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria; Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| | - Andrea Trost
- Research Program for Experimental Ophthalmology and Glaucoma Research, Department of Ophthalmology and Optometry, University Hospital of the Paracelsus Medical University Salzburg, Muellner Hauptstr. 48, 5020 Salzburg, Austria.
| |
Collapse
|
23
|
Han X, Yan T, Wang L, He B, Yu H. Knockdown of PTEN promotes colon cancer progression and induces M2 macrophage polarization in the colon cancer cell environment. INDIAN J PATHOL MICR 2023; 66:478-487. [PMID: 37530327 DOI: 10.4103/ijpm.ijpm_786_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Objective This article aims to study the effect of phosphate and tension homolog deleted on chromosome ten (PTEN) knockdown on colon cancer progression and macrophage polarization in the cancer environment. Materials and Methods and Results The expression of PTEN in colon cancer tissues and colon cancer cells was significantly lower than in precancerous tissues or CCD-18Co cells, and the decrease was most evident in SW620 cells. The expressions of phosphate (p)-p38, c-Jun N-terminal kinase (JNK), activator protein 1 (AP-1), B-cell lymphoma-2 (Bcl-2) protein in colon cancer tissues and cells were significantly higher than in precancerous tissues or CCD-18Co cells (P-values < 0.05). Bcl-2-associated X (Bax) and Caspase-3 expressions in colon cancer tissues and cells were significantly lower than in precancerous tissues or CCD-18Co cells (P-values < 0.05). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was applied to measure cell viability. Transwell evaluated the cell migration and invasion ability. Si-PTEN improved the proliferation, migration, and invasion of SW620 cells (P-values < 0.05). The expression levels of arginase-1 (Arg-1), CD163, CD206 in colon cancer tissues were significantly higher than in precancerous tissues (P-values < 0.05). The cell cycle, the number of M1 and M2 double-positive cells were assessed by flow cytometry. Si-PTEN reduced the expression of tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β), and inducible nitric oxide synthase (iNOS), which upregulated the expression of Arg-1, CD206, CD163, p-p38, JNK, and AP-1 (P-values < 0.05). Conclusion Si-PTEN promoted colon cancer progression and induced the polarization of M2 tumor-associated macrophages in the colon cancer cell environment.
Collapse
Affiliation(s)
- Xu Han
- General Surgery Department, The Fourth Hospital of Changsha, Changsha City, Hunan Province, China
| | - Ting Yan
- General Surgery Department, The Fourth Hospital of Changsha, Changsha City, Hunan Province, China
| | - Lina Wang
- Department of General Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou City, Henan Province, China
| | - Bin He
- General Surgery Department, The Fourth Hospital of Changsha, Changsha City, Hunan Province, China
| | - Huaxu Yu
- General Surgery Department, The Fourth Hospital of Changsha, Changsha City, Hunan Province, China
| |
Collapse
|
24
|
Li R, Jia H, Si M, Li X, Ma Z, Zhu Y, Sun W, Zhu F, Luo S. Loureirin B protects against cerebral ischemia/reperfusion injury through modulating M1/M2 microglial polarization via STAT6 / NF-kappaB signaling pathway. Eur J Pharmacol 2023:175860. [PMID: 37331681 DOI: 10.1016/j.ejphar.2023.175860] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The latest research indicates that modulating microglial polarization from M1 to M2 phenotype may be a coping therapy for ischemic stroke. The present study thereby evaluated the effects of loureirin B (LB), a monomer compound extracted from Sanguis Draconis flavones (SDF), on cerebral ischemic injury and the potential mechanisms. The middle cerebral artery occlusion (MCAO) model was established in male Sprague-Dawley rats to induce cerebral ischemia/reperfusion (I/R) injury in vivo, and BV2 cells were exposed to oxygen-glucose deprivation and reintroduction (OGD/R) to mimic cerebral I/R injury in vitro. The results showed that LB significantly reduced infarct volume, neurological deficits and neurobehavioral deficits, apparently improved histopathological changes and neuronal loss in cortex and hippocampus of MCAO/R rats, markedly decreased the proportion of M1 microglia cells and the level of pro-inflammatory cytokines, and increased the proportion of M2 microglia and the level of anti-inflammatory cytokines both in vivo and in vitro. In addition, LB evidently improved the p-STAT6 expression and reduced the NF-κB (p-p65) expression after cerebral I/R injury in vivo and in vitro. IL-4 (a STAT6 agonist) exhibited a similar impact to that of LB, while AS1517499 (a STAT6 inhibitor) significantly reversed the effect of LB on BV-2 cells after OGD/R. These findings point to the protection of LB against cerebral I/R injury by modulating M1/M2 polarization of microglia via the STAT6/NF-κB signaling pathway, hence LB may be a viable treatment option for ischemic stroke.
Collapse
Affiliation(s)
- Rui Li
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Huiyu Jia
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Min Si
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Xinwei Li
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Zheng Ma
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Yu Zhu
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China
| | - Wuyi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui, 230032, China.
| | - Fengqin Zhu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China.
| | - Shengyong Luo
- Anhui Medical College (Anhui Academy of Medical Sciences), Hefei, Anhui, 230061, China.
| |
Collapse
|
25
|
Wang H, Li J, Zhang H, Wang M, Xiao L, Wang Y, Cheng Q. Regulation of microglia polarization after cerebral ischemia. Front Cell Neurosci 2023; 17:1182621. [PMID: 37361996 PMCID: PMC10285223 DOI: 10.3389/fncel.2023.1182621] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Stroke ranks second as a leading cause of death and permanent disability globally. Microglia, innate immune cells in the brain, respond rapidly to ischemic injury, triggering a robust and persistent neuroinflammatory reaction throughout the disease's progression. Neuroinflammation plays a critical role in the mechanism of secondary injury in ischemic stroke and is a significant controllable factor. Microglia activation takes on two general phenotypes: the pro-inflammatory M1 type and the anti-inflammatory M2 type, although the reality is more complex. The regulation of microglia phenotype is crucial to controlling the neuroinflammatory response. This review summarized the key molecules and mechanisms of microglia polarization, function, and phenotypic transformation following cerebral ischemia, with a focus on the influence of autophagy on microglia polarization. The goal is to provide a reference for the development of new targets for the treatment for ischemic stroke treatment based on the regulation of microglia polarization.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jingjing Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Han Zhang
- School of Medicine, Nantong University, Nantong, China
| | - Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lifang Xiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yitong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
26
|
Scott MC, Haase CM, Olson SD, Cox CS. Dexmedetomidine Alters the Inflammatory Profile of Rat Microglia In Vitro. Neurocrit Care 2023; 38:688-697. [PMID: 36418766 PMCID: PMC10754354 DOI: 10.1007/s12028-022-01638-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/27/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Microglia are a primary mediator of the neuroinflammatory response to neurologic injury, such as that in traumatic brain injury. Their response includes changes to their cytokine expression, metabolic profile, and immunophenotype. Dexmedetomidine (DEX) is an α2 adrenergic agonist used as a sedative in critically ill patients, such as those with traumatic brain injury. Given its pharmacologic properties, DEX may alter the phenotype of inflammatory microglia. METHODS Primary microglia were isolated from Sprague-Dawley rats and cultured. Microglia were activated using multiple mediators: lipopolysaccharide (LPS), polyinosinic-polycytidylic acid (Poly I:C), and traumatic brain injury damage-associated molecular patterns (DAMP) from a rat that sustained a prior controlled cortical impact injury. After activation, cultures were treated with DEX. At the 24-h interval, the cell supernatant and cells were collected for the following studies: cytokine expression (tumor necrosis factor-α [TNFα], interleukin-10 [IL-10]) via enzyme-linked immunosorbent assay, 6-phosphofructokinase enzyme activity assay, and immunophenotype profiling with flow cytometry. Cytokine expression and metabolic enzyme activity data were analyzed using two-way analysis of variance. Cell surface marker expression was analyzed using FlowJo software. RESULTS In LPS-treated cultures, DEX treatment decreased the expression of TNFα from microglia (mean difference = 121.5 ± 15.96 pg/mL; p < 0.0001). Overall, DEX-treated cultures had a lower expression of IL-10 than nontreated cultures (mean difference = 39.33 ± 14.50 pg/mL, p < 0.0001). DEX decreased IL-10 expression in LPS-stimulated microglia (mean difference = 74.93 ± 12.50 pg/mL, p = 0.0039) and Poly I:C-stimulated microglia (mean difference = 23.27 ± 6.405 pg/mL, p = 0.0221). In DAMP-stimulated microglia, DEX decreased the activity of 6-phosphofructokinase (mean difference = 18.79 ± 6.508 units/mL; p = 0.0421). The microglial immunophenotype was altered to varying degrees with different inflammatory stimuli and DEX treatment. CONCLUSIONS DEX may alter the neuroinflammatory response of microglia. By altering the microglial profile, DEX may affect the progression of neurologic injury.
Collapse
Affiliation(s)
- Michael C Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.3600, Houston, TX, USA.
| | - Candice M Haase
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.3600, Houston, TX, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.3600, Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, 1881 East Road, 3SCR6.3600, Houston, TX, USA
| |
Collapse
|
27
|
Zhang Q, Zhang S, Chen H, Chen G, Cui C, Zhang J, Wang W, Zhang Q, Guo S. Targeting of MALT1 May Improve Functional Recovery and Attenuate Microglia M1 Polarization-Mediated Neuroinflammation During Spinal Cord Injury. Mol Neurobiol 2023; 60:2632-2643. [PMID: 36692707 DOI: 10.1007/s12035-023-03208-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/02/2023] [Indexed: 01/25/2023]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) is involved in neural injury, neuroinflammation, microglia activation, and polarization, while its function in spinal cord injury (SCI) remains unclear. Thus, this study aimed to evaluate the role of MALT1 modification on SCI recovery and its underlying mechanism. SCI surgery or sham surgery was performed in Sprague-Dawley rats. Then, MALT1 knockdown or negative control lentivirus was injected into SCI rats. Subsequently, MALT1 expression, locomotor capability, neural injury, markers for microglia activation and polarization, inflammatory cytokine expressions, and nuclear factor (NF)-κB pathway were detected. SCI rats exhibited higher MALT1 expression, microglia activation and M1 polarization, neuroinflammation, and NF-κB pathway activation, while worse locomotor capacity compared to sham rats (all P < 0.05). In SCI rats, MALT1 knockdown alleviated Basso, Beattie, and Bresnahan score from 10 to 28 days and attenuated HE staining reflected neural injury (all P < 0.05). Besides, MALT1 knockdown declined the number of IBA1+ cells, IBA1+ iNOS+ cells, and IBA1+ CD86+ cells, while enhanced the number of IBA1+ Arg1+ cells and IBA1+ CD206+ cells in SCI rats (all P < 0.05). Meanwhile, MALT1 knockdown declined the expressions of IL-1β, IL-6, and TNF-α in SCI (all P < 0.05), but did not affect IL-10 expression (P > 0.05). Furthermore, MALT1 knockdown suppressed NF-κB pathway activation validated by immunofluorescence staining and western blot assays (all P < 0.05). MALT1 knockdown improves functional recovery, attenuates microglia activation, M1 polarization, and neuroinflammation via inhibiting NF-κB pathway in SCI.
Collapse
Affiliation(s)
- Qingping Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Xi'an Yanta West Road, Xi'an, 710061, Shaanxi, China.
- Department of Neurosurgery, The 6th Affiliated Hospital of Shenzhen University Medical School (Shenzhen Nanshan People's Hospital), No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| | - Shitao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Xi'an Yanta West Road, Xi'an, 710061, Shaanxi, China
- Department of Neurosurgery, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, 710018, Shaanxi, China
| | - Hongquan Chen
- Department of Neurosurgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, Guangdong, China
| | - Gang Chen
- Department of Spine, Xiangtan Central Hospital, Xiangtan, 411100, Hunan, China
| | - Chunhong Cui
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Department of Biology and Academy for Advanced Interdisciplinary Studies, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Junxin Zhang
- Department of Neurosurgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, Guangdong, China
| | - Weiming Wang
- Department of Neurosurgery, Southern University of Science and Technology Hospital, Shenzhen, 518055, Guangdong, China
| | - Qinghua Zhang
- Department of Neurosurgery, The 6th Affiliated Hospital of Shenzhen University Medical School (Shenzhen Nanshan People's Hospital), No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Shiwen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Xi'an Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
28
|
Ning L, Ye N, Ye B, Miao Z, Cao T, Lu W, Xu D, Tan C, Xu Y, Yan J. Qingre Xingyu recipe exerts inhibiting effects on ulcerative colitis development by inhibiting TNFα/NLRP3/Caspase-1/IL-1β pathway and macrophage M1 polarization. Cell Death Discov 2023; 9:84. [PMID: 36890151 PMCID: PMC9995513 DOI: 10.1038/s41420-023-01361-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 03/10/2023] Open
Abstract
As a chronic inflammatory bowel disease, ulcerative colitis (UC) imposes a significant burden on public healthcare worldwide due to its increasing morbidity. Chinese medicines are regarded as potent therapeutic agents for UC treatment with minimal side effects. In the present study, we sought to determine the novel role of a traditional medicine Qingre Xingyu (QRXY) recipe in the development of UC and aimed to contribute to the currently available knowledge about UC by exploring the downstream mechanism of QRXY recipe in UC. Mouse models of UC were established by injections with dextran sulphate sodium (DSS), where the expression of tumor necrosis factor-alpha (TNFα), NLR family pyrin domain containing 3 (NLRP3), and interleukin-1β (IL-1β) was determined followed by an analysis of their interactions. The DSS-treated NLRP3 knockout (-/-) Caco-2 cell model was successfully constructed. The in vitro and in vivo effects of the QRXY recipe on UC were investigated with the determination of disease activity index (DAI), histopathological scores, transepithelial electrical resistance, FITC-dextran, as well as cell proliferation and apoptosis. In vivo and in vitro experiments indicated that the QRXY recipe reduced the degree of intestinal mucosal injury of UC mice and functional damage of DSS-induced Caco-2 cells by inhibition of the TNFα/NLRP3/caspase-1/IL-1β pathway and M1 polarization of macrophages, and TNFα overexpression or NLRP3 knockdown could counterweigh the therapeutic effects of QRXY recipe. To conclude, our study elicited that QRXY inhibited the expression of TNFα and inactivated the NLRP3/Caspase-1/IL-1β pathway, thereby alleviating intestinal mucosal injury and relieving UC in mice.
Collapse
Affiliation(s)
- Liqin Ning
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Ningyuan Ye
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Bai Ye
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, 215600, P. R. China
| | - Tingting Cao
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Weimin Lu
- Department of Internal Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Danhua Xu
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Chang Tan
- Chinese Medicine Master Studio, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China
| | - Yi Xu
- Department of Gastroenterology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, P. R. China.
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China.
| |
Collapse
|
29
|
Xu J, Chen TY, Tai CH, Hsu SH. Bioactive self-healing hydrogel based on tannic acid modified gold nano-crosslinker as an injectable brain implant for treating Parkinson's disease. Biomater Res 2023; 27:8. [PMID: 36755333 PMCID: PMC9909866 DOI: 10.1186/s40824-023-00347-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is one of the most common long-term neurodegenerative diseases. Current treatments for PD are mostly based on surgery and medication because of the limitation and challenges in selecting proper biomaterials. In this study, an injectable bioactive hydrogel based on novel tannic acid crosslinker was developed to treat PD. METHODS The oxidized tannic acid modified gold nano-crosslinker was synthesized and used to effectively crosslink chitosan for preparation of the bioactive self-healing hydrogel. The crosslinking density, conductivity, self-healing ability, and injectability of the hydrogel were characterized. Abilities of the hydrogel to promote the proliferation and differentiation of neural stem cells (NSCs) were assessed in vitro. Anti-inflammatory property was analyzed on J774A.1 macrophages. The hydrogel was injected in the PD rat model for evaluation of the motor function recovery, electrophysiological performance improvement, and histological repair. RESULTS The hydrogel exhibited self-healing property and 34G (~ 80 μm) needle injectability. NSCs grown in the hydrogel displayed long-term proliferation and differentiation toward neurons in vitro. Besides, the hydrogel owned strong anti-inflammatory and antioxidative capabilities to rescue inflamed NSCs (~ 90%). Brain injection of the bioactive hydrogel recovered the motor function of PD rats. Electrophysiological measurements showed evident alleviation of irregular discharge of nerve cells in the subthalamic nucleus of PD rats administered with the hydrogel. Histological examination confirmed that the hydrogel alone significantly increased the density of tyrosine hydroxylase positive neurons and fibers as well as reduced inflammation, with a high efficacy similar to drug-loaded hydrogel. CONCLUSION The new bioactive hydrogel serves as an effective brain injectable implant to treat PD and a promising biomaterial for developing novel strategies to treat brain diseases.
Collapse
Affiliation(s)
- Junpeng Xu
- grid.19188.390000 0004 0546 0241Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617 Taiwan, Republic of China
| | - Tsai-Yu Chen
- grid.19188.390000 0004 0546 0241Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617 Taiwan, Republic of China
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, No.7, Zhongshan South Road, Zhongzheng District, Taipei, 100225, Taiwan, Republic of China.
| | - Shan-hui Hsu
- grid.19188.390000 0004 0546 0241Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617 Taiwan, Republic of China ,grid.59784.370000000406229172Institute of Cellular and System Medicine, National Health Research Institutes, No. 35 Keyan Road, Miaoli, 35053 Taiwan, Republic of China
| |
Collapse
|
30
|
Cheng Z, Li X, Ye X, Yu R, Deng Y. Purpurogallin Reverses Neuronal Apoptosis and Enhances "M2" Polarization of Microglia Under Ischemia via Mediating the miR-124-3p/TRAF6/NF-κB Axis. Neurochem Res 2023; 48:375-392. [PMID: 36131212 DOI: 10.1007/s11064-022-03752-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 02/04/2023]
Abstract
Purpurogallin (PPG) has been demonstrated to exert an anti-inflammatory function in neurological diseases. This study aimed at investigating the role of PPG on microglial polarization post ischemic stroke as well as the underlying mechanism. Mouse hippocampal neurons HT-22 and microglial BV2 cells were treated by oxygen and glucose deprivation to simulate an in-vitro ischemia model. qRT-PCR and ELISA examined expression of cytokines in microglia. CCK8 and flow cytometry measured HT-22 cell viability and apoptosis, respectively. The levels of miR-124-3p and TRAF6/NF-κB were determined. A mouse cerebral ischemia model was set up using middle cerebral artery occlusion (MCAO) method. After being dealt with PPG, the neurological functions, brain edema, neuronal apoptosis, and microglia activation of the mice were evaluated. As suggested by the results, PPG transformed "M1" to "M2" polarization of BV2 cells, and abated HT-22 cell apoptosis. PPG enhanced the neurological functions, alleviated brain edema, and decreased neuroinflammatory responses, and neuronal apoptosis in the brain lesions of MCAO mice. Furthermore, PPG enhanced miR-124-3p and repressed the TRAF6/NF-κB pathway. miR-124-3p suppressed the TRAF6/NF-κB pathway by targeting TRAF6. Collectively, PPG alleviates ischemia-induced neuronal damage and microglial inflammation by modulating the miR-124-3p/TRAF6/NF-κB pathway.
Collapse
Affiliation(s)
- Zongxin Cheng
- Department of Neurology, Nanchang First Hospital, No.128 Xiangshan North Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| | - Xinming Li
- Department of Neurology, Nanchang First Hospital, No.128 Xiangshan North Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Xiaohua Ye
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Rong Yu
- Department of Neurology, Nanchang First Hospital, No.128 Xiangshan North Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Youqing Deng
- Department of Neurology, Nanchang First Hospital, No.128 Xiangshan North Road, Donghu District, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
31
|
Hu Y, Yin J, Yang G. Melatonin upregulates BMAL1 to attenuate chronic sleep deprivation-related cognitive impairment by alleviating oxidative stress. Brain Behav 2023; 13:e2836. [PMID: 36563187 PMCID: PMC9847595 DOI: 10.1002/brb3.2836] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/07/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To investigate the mechanism underlying the regulatory effect of melatonin on chronic sleep deprivation-related cognitive impairment. METHODS Chronic sleep deprivation (CSD) model was established using the MMPM method. After the model was established, melatonin receptor agonist and inhibitor were given, respectively. Water maze was conducted to record the escape latency and the duration of crossing the platform of space exploration. The concentration of TNF-α, IL-6, MDA, and SOD was measured by ELISA. Immunofluorescence was used to determine the expression level of CD86 and CD206, while the mRNA expression of Bax, Bcl-2, P65, IκB, and BMAL1 was detected by qPCR. Western blotting assay was utilized to determine the protein expression of Bax, Bcl-2, P65, p-P65, IκB, p-I κB, and BMAL1. RESULTS Compared with the control, the escape latency was greatly increased on the second and third day, accompanied by the increased expression of TNF-α, IL-6, MDA, and SOD in serum. Furthermore, dramatically upregulated Bax, Bcl-2, P65, IκB, and CD86 were observed in the model group, accompanied by the declined expression level of BMAL1 and CD206. Compared with the model group, the escape latency was declined, the concentration of TNF-α, IL-6, MDA, and SOD was decreased, the expression level of Bax, Bcl-2, P65, IκB, and CD86 was declined, and the level of BMAL1 and CD206 was promoted by the treatment of the melatonin agonist, while the opposite results were observed under the treatment of the melatonin inhibitor. CONCLUSION Melatonin upregulates BMAL1 to attenuate chronic sleep deprivation-related cognitive impairment by alleviating oxidative stress.
Collapse
Affiliation(s)
- Yujie Hu
- Department of Neurology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Jierong Yin
- Department of Neurology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| | - Guoshuai Yang
- Department of Neurology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital, Haikou, China
| |
Collapse
|
32
|
Yu LH, Jia GW, Liu YL, Wang SR, Ma JX. Vagus nerve stimulation is a potential treatment for ischemic stroke. Neural Regen Res 2023; 18:825-831. [DOI: 10.4103/1673-5374.350698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
33
|
Yu Z, Su G, Zhang L, Liu G, Zhou Y, Fang S, Zhang Q, Wang T, Huang C, Huang Z, Li L. Icaritin inhibits neuroinflammation in a rat cerebral ischemia model by regulating microglial polarization through the GPER-ERK-NF-κB signaling pathway. Mol Med 2022; 28:142. [PMID: 36447154 PMCID: PMC9706854 DOI: 10.1186/s10020-022-00573-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/09/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Activated microglia play a key role in initiating the inflammatory cascade following ischemic stroke and exert proinflammatory or anti-inflammatory effects, depending on whether they are polarized toward the M1 or M2 phenotype. The present study investigated the regulatory effect of icaritin (ICT) on microglial polarization in rats after cerebral ischemia/reperfusion injury (CI/RI) and explored the possible anti-inflammatory mechanisms of ICT. METHODS A rat model of transient middle cerebral artery occlusion (tMCAO) was established. Following treatment with ICT, a G protein-coupled estrogen receptor (GPER) inhibitor or an extracellular signal-regulated kinase (ERK) inhibitor, the Garcia scale and rotarod test were used to assess neurological and locomotor function. 2,3,5-Triphenyltetrazolium chloride (TTC) and Fluoro-Jade C (FJC) staining were used to evaluate the infarct volume and neuronal death. The levels of inflammatory factors in the ischemic penumbra were evaluated using enzyme-linked immunosorbent assays (ELISAs). In addition, western blotting, immunofluorescence staining and quantitative PCR (qPCR) were performed to measure the expression levels of markers of different microglial phenotypes and proteins related to the GPER-ERK-nuclear factor kappa B (NF-κB) signaling pathway. RESULTS ICT treatment significantly decreased the cerebral infarct volume, brain water content and fluorescence intensity of FJC; improved the Garcia score; increased the latency to fall and rotation speed in the rotarod test; decreased the levels of interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), Iba1, CD40, CD68 and p-P65-NF-κB; and increased the levels of CD206 and p-ERK. U0126 (an inhibitor of ERK) and G15 (a selective antagonist of GPER) antagonized these effects. CONCLUSIONS These findings indicate that ICT plays roles in inhibiting the inflammatory response and achieving neuroprotection by regulating GPER-ERK-NF-κB signaling and then inhibiting microglial activation and M1 polarization while promoting M2 polarization, which provides a new therapeutic for against cerebral ischemic stroke.
Collapse
Affiliation(s)
- Zining Yu
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Graduate School, Gannan Medical University, Ganzhou, 341000 China
| | - Guangjun Su
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Graduate School, Gannan Medical University, Ganzhou, 341000 China
| | - Limei Zhang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000 China
| | - Gaigai Liu
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Graduate School, Gannan Medical University, Ganzhou, 341000 China
| | - Yonggang Zhou
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000 China
| | - Shicai Fang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Graduate School, Gannan Medical University, Ganzhou, 341000 China
| | - Qian Zhang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Graduate School, Gannan Medical University, Ganzhou, 341000 China
| | - Tianyun Wang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000 China
| | - Cheng Huang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Institute for Medical Sciences of Pain, Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000 China
| | - Zhihua Huang
- grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Institute for Medical Sciences of Pain, Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000 China
| | - Liangdong Li
- grid.452437.3First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000 China ,grid.440714.20000 0004 1797 9454Ganzhou Key Laboratory of Neuroinflammation Research, Gannan Medical University, Ganzhou, 341000 China
| |
Collapse
|
34
|
Zhang D, Chang R, Ren Y, He Y, Guo S, Guan F, Yao M. Injectable and reactive oxygen species-scavenging gelatin hydrogel promotes neural repair in experimental traumatic brain injury. Int J Biol Macromol 2022; 219:844-863. [PMID: 35961554 DOI: 10.1016/j.ijbiomac.2022.08.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/06/2022] [Indexed: 12/26/2022]
Abstract
Oxidative stress caused by the overexpression of reactive oxygen species (ROS) plays an important role in the pathogenesis of traumatic brain injury (TBI). Accumulation of ROS can lead to cell death, neurodegeneration, and neurological deficit. Therefore, the design and application of functional materials with ROS scavenging ability is of great significance for neural repair. Herein, an injectable and antioxidant hydrogel was developed for TBI treatment based on the Schiff base reaction of gallic acid-conjugated gelatin (GGA) and oxidized dextran (Odex). The resulting GGA/Odex hydrogel could effectively scavenge DPPH and ABTS radicals, as well as protect cells from the oxidative damage in vitro. Moreover, GGA/Odex hydrogel possessed well biocompatible features. In a moderate TBI mouse model, in situ implantation of GGA6Odex hydrogel efficiently facilitated neurogenesis and promoted the motor, learning and memory abilities. Also, this composite hydrogel suppressed oxidative stress and inflammation via the activation of Nrf2/HO-1 pathway and the regulating of inflammatory factors secretion and macrophage/microglia polarization. Therefore, this injectable and ROS-scavenging GGA6Odex hydrogel is a promising biomaterial for tissue regenerative medicine, including TBI and other tissue repair relevant to raised ROS circumstance.
Collapse
Affiliation(s)
- Dan Zhang
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Rong Chang
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Yikun Ren
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Yuanmeng He
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Shen Guo
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China
| | - Fangxia Guan
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China.
| | - Minghao Yao
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou 450001, PR China.
| |
Collapse
|
35
|
Wu W, Zhang X, Wang S, Li T, Hao Q, Li S, Yao W, Sun R. Pharmacological inhibition of the cGAS-STING signaling pathway suppresses microglial M1-polarization in the spinal cord and attenuates neuropathic pain. Neuropharmacology 2022; 217:109206. [PMID: 35926582 DOI: 10.1016/j.neuropharm.2022.109206] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/14/2022] [Accepted: 07/23/2022] [Indexed: 12/23/2022]
Abstract
Neuroinflammation plays a vital role in the development of neuropathic pain and is mediated mainly by microglia. Suppressing microglial M1-polarization attenuates neuropathic pain. Recently, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has emerged as a key mediator of inflammation and shows potential in modulating microglial polarization. In this study, we evaluated whether cGAS-STING is a potential therapeutic target. Spared nerve injury (SNI) surgery was conducted in adult male rats to establish a neuropathic pain model. We showed that SNI promoted microglial M1-polarization and induced cGAS-STING pathway activation in the spinal cord. Double-label immunofluorescence assays showed that cGAS-STING activation mainly occurred in neurons and microglia but not astrocytes. We further conducted in vitro experiments using BV-2 microglial cells. The results showed that LPS-induced microglial M1-polarization was accompanied by cGAS-STING pathway activation, but cGAS-STING inhibition by antagonists suppressed LPS-induced microglial M1-polarization. In vivo, we also showed that a cGAS antagonist and a STING antagonist suppressed the microglial M1-polarization and ameliorated the mechanical allodynia induced by SNI. These findings suggested that the cGAS-STING pathway might be a potential therapeutic target for treating neuropathic pain. However, further research is warranted to verify our findings in female rodents.
Collapse
Affiliation(s)
- Wenyao Wu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianwei Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuo Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quanshui Hao
- Department of Anesthesiology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenlong Yao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rao Sun
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Park JS, Kim MS, Joung MY, Park HJ, Ho MJ, Choi JH, Seo JH, Song WH, Choi YW, Lee S, Choi YS, Kang MJ. Design of Montelukast Nanocrystalline Suspension for Parenteral Prolonged Delivery. Int J Nanomedicine 2022; 17:3673-3690. [PMID: 36046838 PMCID: PMC9423109 DOI: 10.2147/ijn.s375888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Montelukast (MTK), a representative leukotriene receptor antagonist, is currently being investigated as a potential candidate for treating Alzheimer’s disease. For potent and effective dosing in elderly patients, a parenteral prolonged delivery system is favored, with improved medication adherence with reduced dosage frequency. Purpose This study aimed to design a nanocrystalline suspension (NS)-based MTK prolonged delivery system and evaluate its pharmacokinetics profile and local tolerability following subcutaneous administration. Methods To decelerate the dissolution rate, the amorphous MTK raw material was transformed into a crystalline state using a solvent-mediated transformation method and subsequently formulated into NS using a bead-milling technique. The MTK NSs were characterized by morphology, particle size, crystallinity, and in vitro dissolution profiles. The pharmacokinetic profile and local tolerability at the injection site following subcutaneous injection of MTK suspension were evaluated in rats. Results Microscopic and physical characterization revealed that the amorphous MTK powder was lucratively transformed into a crystalline form in acidic media (pH 4). MTK crystalline suspensions with different diameters (200 nm, 500 nm, and 3 μm) were uniformly prepared using bead-milling technology, employing polysorbate 80 as suspending agent. Prepared crystalline suspensions exhibited analogous crystallinity (melting point, 150°C) and size-dependent in vitro dissolution profiles. MTK NSs with particle sizes of 200 nm and 500 nm provided a protracted pharmacokinetic profile for up to 4 weeks in rats, with a higher maximum drug concentration in plasma than the 3 μm-sized injectable suspensions. Histopathological examination revealed that MTK NS caused chronic granulomatous inflammation at the injection site, which resolved after 4 weeks. Conclusion The MTK parenteral NS delivery system is expected to be a valuable tool for treating Alzheimer’s disease with extended dose intervals.
Collapse
Affiliation(s)
- Jun Soo Park
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Min Seop Kim
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Min Yeong Joung
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Hyun Jin Park
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Myoung-Jin Ho
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Jun Hyuk Choi
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Jae Hee Seo
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Woo Heon Song
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Young Wook Choi
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Sangkil Lee
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Yong Seok Choi
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Myung Joo Kang
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
37
|
Xia W, Xu Y, Gong Y, Cheng X, Yu T, Yu G. Microglia Involves in the Immune Inflammatory Response of Poststroke Depression: A Review of Evidence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2049371. [PMID: 35958023 PMCID: PMC9363171 DOI: 10.1155/2022/2049371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
Poststroke depression (PSD) does not exist before and occurs after the stroke. PSD can appear shortly after the onset of stroke or be observed in the weeks and months after the acute or subacute phase of stroke. The pathogenesis of PSD is unclear, resulting in poor treatment effects. With research advancement, immunoactive cells in the central nervous system, particularly microglia, play a role in the occurrence and development of PSD. Microglia affects the homeostasis of the central nervous system through various factors, leading to the occurrence of depression. The research progress of microglia in PSD has been summarized to review the evidence regarding the pathogenesis and treatment target of PSD in the future.
Collapse
Affiliation(s)
- Weili Xia
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
| | - Yong Xu
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
| | - Yuandong Gong
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
| | - Xiaojing Cheng
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
| | - Tiangui Yu
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
| | - Gongchang Yu
- Shandong Mental Health Center, Shandong University, Jinan, Shandong 250014, China
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| |
Collapse
|
38
|
Yang QY, Li XW, Yang R, Qin TY, Long H, Zhang SB, Zhang F. Effects of intraperitoneal injection of lipopolysaccharide-induced peripheral inflammation on dopamine neuron damage in rat midbrain. CNS Neurosci Ther 2022; 28:1624-1636. [PMID: 35789066 PMCID: PMC9437226 DOI: 10.1111/cns.13906] [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: 03/04/2022] [Revised: 05/24/2022] [Accepted: 06/04/2022] [Indexed: 11/26/2022] Open
Abstract
Introduction Current studies have documented neuroinflammation is implicated in Parkinson's disease. Recently, growing evidence indicated peripheral inflammation plays an important role in regulation of neuroinflammation and thus conferring protection against dopamine (DA) neuronal damage. However, the underlying mechanisms are not clearly illuminated. Methods The effects of intraperitoneal injection of LPS (LPS[i.p.])‐induced peripheral inflammation on substantia nigra (SN) injection of LPS (LPS[SN])‐elicited DA neuronal damage in rat midbrain were investigated. Rats were intraperitoneally injected with LPS (0.5 mg/kg) daily for 4 consecutive days and then given single injection of LPS (8 μg) into SN with an interval of 0 (LPS(i.p.) 0 day ± LPS(SN)), 30 (LPS(i.p.) 30 days ± LPS(SN)), and 90 (LPS(i.p.) 90 days ± LPS(SN)) days after LPS(i.p.) administration. Results LPS(i.p.) increased the levels of inflammatory factors in peripheral blood in (LPS(i.p.) 0 day ± LPS(SN)). Importantly, in (LPS(i.p.) 0 day ± LPS(SN)) and (LPS(i.p.) 30 days ± LPS(SN)), LPS(i.p.) attenuated LPS(SN)‐induced DA neuronal loss in SN. Besides, LPS(i.p.) reduced LPS(SN)‐induced microglia and astrocytes activation in SN. Furtherly, LPS(i.p.) reduced pro‐inflammatory M1 microglia markers mRNA levels and increased anti‐inflammatory M2 microglia markers mRNA levels. In addition, the increased T‐cell marker expression and the decreased M1 microglia marker expression and more DA neuronal survival were discerned at the same area of rat midbrain in LPS(SN)‐induced DA neuronal damage 30 days after LPS(i.p.) application. Conclusion This study suggested LPS(i.p.)‐induced peripheral inflammation might cause T cells to infiltrate the brain to regulate microglia‐mediated neuroinflammation, thereby protecting DA neurons.
Collapse
Affiliation(s)
- Qiu-Yu Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Xian-Wei Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Rong Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Ting-Yang Qin
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Hong Long
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Shi-Bin Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, China
| |
Collapse
|
39
|
Carion TW, Wang Y, Stambersky A, Ebrahim AS, Berger EA. A Dual Role for Cysteinyl Leukotriene Receptors in the Pathogenesis of Corneal Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2331-2342. [PMID: 35470258 PMCID: PMC9117469 DOI: 10.4049/jimmunol.2100474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 03/12/2022] [Indexed: 05/17/2023]
Abstract
Cysteinyl leukotrienes (CysLTs) have been defined as central mediators of inflammation. Despite our extensive understanding of these bioactive lipid mediators in the pathogenesis of diseases such as asthma, allergic rhinitis, and even neurological disorders, information regarding the eye is markedly lacking. As a result, this study examined the expression profiles of two major CysLT receptors, CysLT1 and CysLT2, in the cornea using experimental mouse models of Pseudomonas aeruginosa-induced keratitis with contrasting outcomes: susceptible C57BL/6 (B6) and resistant BALB/c. Postinfection, disparate levels of CysLT receptors were accompanied by distinct expression profiles for select proinflammatory and anti-inflammatory cell surface markers detected on macrophages and polymorphonuclear neutrophils between the two strains. Further, inhibition of either CysLT receptor converted the disease response of both strains, where corneal perforation was prevented in B6 mice, and BALB/c mice fared significantly worse. In addition, receptor antagonist studies revealed changes in inflammatory cell infiltrate phenotypes and an influence on downstream CysLT receptor signaling pathways. Although the B6 mouse model highlights the established proinflammatory activities related to CysLT receptor activation, results generated from BALB/c mice indicate a protective mechanism that may be essential to disease resolution. Further, basal expression levels of CysLT1 and CysLT2 were significantly higher in uninfected corneas of both mouse strains as opposed to during infection, suggestive of a novel role in homeostatic maintenance within the eye. In light of these findings, therapeutic targeting of CysLT receptors extends beyond inhibition of proinflammatory activities and may impact inflammation resolution, as well as corneal surface homeostasis.
Collapse
Affiliation(s)
- Thomas W Carion
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Yuxin Wang
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Ashten Stambersky
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Abdul Shukkur Ebrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| | - Elizabeth A Berger
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
40
|
Liu G, Li M, Qian S, Yu L, Qian L, Feng X. Interleukin-35 exhibits protective effects in a rat model of hypoxic-ischemic encephalopathy through the inhibition of microglia-mediated inflammation. Transl Pediatr 2022; 11:651-662. [PMID: 35685068 PMCID: PMC9173876 DOI: 10.21037/tp-22-100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/13/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) brain damage is related to inflammatory responses and oxidative stress. Interleukin (IL)-35 is an antioxidant and anti-inflammatory cytokine. Thus, the effect of IL-35 treatment on neonatal rats with hypoxic-ischemic brain injury was investigated. METHODS A total of 96 7-day-old Sprague Dawley rats were randomly divided into three groups: sham group, HIE group, and IL-35 group. After left common carotid occlusion and 2.5 h hypoxia (HI injury), IL-35 (20 µg/g) was intraperitoneally (i.p.) administered to the pups. In vitro, BV2 cells were treated with or without IL-35 6 h before oxygen-glucose deprivation (OGD) insult and the microglia culture medium (MCM) was co-cultured with b.End3 cerebral vascular endothelial cells. Microglial polarization and activation were assessed by real-time quantitative polymerase chain reaction (RT-qPCR), Western blot, and enzyme-linked immunosorbent assay (ELISA). Endothelial cell dysfunction was measured by cell counting kit-8 and Western blot assays. RESULTS Administration of IL-35 alleviated neurological deficiencies, decreased brain edema, ameliorated cerebral infarction, and limited M1 microglial polarization in HI-injured pups. Meanwhile, IL-35 decreased pro-inflammatory cytokines, tumor necrosis factor-α, IL-1β, and reactive oxygen species generation in OGD-induced bEnd.3 cells. Furthermore, IL-35 treatment could reverse the vascular endothelial cell injury induced by microglial polarization. Finally, IL-35 markedly suppressed the activation of hypoxia-inducible factor-1α (HIF-1α) and the nuclear factor-κB (NF-κB) signaling pathway in vivo and in vitro. CONCLUSIONS IL-35 relieved hypoxic-ischemic-induced brain injury and inhibited the inflammatory response by suppressing microglial polarization and activation. These results suggest that IL-35 might have potential applications for the treatment of HIE.
Collapse
Affiliation(s)
- Guangliang Liu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China.,Department of Pediatrics, Binhai County People's Hospital, Bianhai, China
| | - Ming Li
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, China
| | - Shuang Qian
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lulu Yu
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, China
| | - Lei Qian
- Department of Laboratory Medicine, Binhai County People's Hospital, Bianhai, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
41
|
Dai Y, Yan M, Wan J, Xiao T. Maf1 mitigates sevoflurane-induced microglial inflammatory damage and attenuates microglia-mediated neurotoxicity in HT-22 cells by activating the AMPK/Nrf2 signaling. Neurotoxicology 2022; 90:237-245. [PMID: 35430185 DOI: 10.1016/j.neuro.2022.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 02/26/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Maf1 has been found to play protective function against neuroinflammation and neuroapoptosis. This study seeks to explore whether and how Maf1 is involved in sevoflurane (Sev)-induced neuroinflammation and microglia-mediated neurotoxicity. METHODS qRT-PCR and western blot were used to detect the gene expression. ELISA was used to detect inflammatory factors. Cell viability was evaluated by using the Cell Counting Kit-8 kit. Neuroapoptosis was assessed with trhe Caspase-3 Assay Kit and the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) technique. RESULTS Maf1 expression was downregulated in Sev-stimulated BV2 microglial cells. Maf1 overexpression down-regulates the expression of pro-inflammatory M1-type markers (CD86, iNOS, IFN-γ) and up-regulates the expression of anti-inflammatory M2-type markers (CD206, TGF-β, Arg-1), and Maf1 reduces the Sev-induced inflammatory response in BV2 cells. After Maf1 overexpression, the relative expression of p-AMPK/AMPK and nucleus-Nrf2 increased significantly in BV2 cells treated with Sev. Inhibition of AMPK/Nrf2 pathway by compound C reverses anti-inflammatory effect of Maf1 in Sev-stimulated BV2 cells. Compound C reverses the effect of Maf1 on microglia-mediated neurotoxicity in HT-22 hippocampal neuronal cells. CONCLUSIONS Maf1 mitigates Sev-induced microglial inflammatory damage and attenuates microglia-mediated neurotoxicity by activating the AMPK/Nrf2 signaling.
Collapse
Affiliation(s)
- Yunyi Dai
- Department of Neurology, The First People's Hospital of Shangqiu, China
| | - Mingguang Yan
- Department of Laboratory Medicine, The First People's Hospital of Shangqiu, China
| | - Juan Wan
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, China
| | - Tao Xiao
- Department of Neurosurgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, China.
| |
Collapse
|
42
|
Attaluri S, Upadhya R, Kodali M, Madhu LN, Upadhya D, Shuai B, Shetty AK. Brain-Specific Increase in Leukotriene Signaling Accompanies Chronic Neuroinflammation and Cognitive Impairment in a Model of Gulf War Illness. Front Immunol 2022; 13:853000. [PMID: 35572589 PMCID: PMC9099214 DOI: 10.3389/fimmu.2022.853000] [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: 01/11/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Persistent cognitive impairment is a primary central nervous system-related symptom in veterans afflicted with chronic Gulf War Illness (GWI). Previous studies in a rat model have revealed that cognitive dysfunction in chronic GWI is associated with neuroinflammation, typified by astrocyte hypertrophy, activated microglia, and enhanced proinflammatory cytokine levels. Studies in a mouse model of GWI have also shown upregulation of several phospholipids that serve as reservoirs of arachidonic acid, a precursor of leukotrienes (LTs). However, it is unknown whether altered LT signaling is a component of chronic neuroinflammatory conditions in GWI. Therefore, this study investigated changes in LT signaling in the brain of rats displaying significant cognitive impairments six months after exposure to GWI-related chemicals and moderate stress. The concentration of cysteinyl LTs (CysLTs), LTB4, and 5-Lipoxygenase (5-LOX), the synthesizing enzyme of LTs, were evaluated. CysLT and LTB4 concentrations were elevated in the hippocampus and the cerebral cortex, along with enhanced 5-LOX expression in neurons and microglia. Such changes were also associated with increased proinflammatory cytokine levels in the hippocampus and the cerebral cortex. Enhanced CysLT and LTB4 levels in the brain could also be gleaned from their concentrations in brain-derived extracellular vesicles in the circulating blood. The circulating blood in GWI rats displayed elevated proinflammatory cytokines with no alterations in CysLT and LTB4 concentrations. The results provide new evidence that a brain-specific increase in LT signaling is another adverse alteration that potentially contributes to the maintenance of chronic neuroinflammation in GWI. Therefore, drugs capable of modulating LT signaling may reduce neuroinflammation and improve cognitive function in GWI. Additional findings demonstrate that altered LT levels in the brain could be tracked efficiently by analyzing brain-derived EVs in the circulating blood.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX, United States
| |
Collapse
|
43
|
Zileuton, a 5-Lipoxygenase Inhibitor, Attenuates Haemolysate-Induced BV-2 Cell Activation by Suppressing the MyD88/NF-κB Pathway. Int J Mol Sci 2022; 23:ijms23094910. [PMID: 35563304 PMCID: PMC9104905 DOI: 10.3390/ijms23094910] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
M1 microglia induce neuroinflammation-related neuronal death in animal models of spontaneous subarachnoid haemorrhage. Zileuton is a 5-lipoxygenase inhibitor that reduces the levels of downstream pro-inflammatory cytokines. This study aimed to investigate whether zileuton inhibits microglial activation and describe its underlying mechanisms. BV-2 cells were exposed to 1 mg/mL haemolysate for 30 min, followed by treatment with different concentrations (5, 10, 15, or 20 μM) of zileuton for 24 h. The cells were then assessed for viability, polarisation, and protein expression levels. Haemolysate increases the viability of BV-2 cells and induces M1 polarisation. Subsequent exposure to high concentrations of zileuton decreased the viability of BV-2 cells, shifted the polarisation to the M2 phenotype, suppressed the expression of 5-lipoxygenase, decreased tumour necrosis factor α levels, and increased interleukin-10 levels. Furthermore, high concentrations of zileuton suppressed the expression of myeloid differentiation primary response protein 88 and reduced the phosphorylated-nuclear factor-kappa B (NF-kB)/NF-kB ratio. Therefore, phenotype reversal from M1 to M2 is a possible mechanism by which zileuton attenuates haemolysate-induced neuroinflammation after spontaneous subarachnoid haemorrhage.
Collapse
|
44
|
Vats A, Xi Y, Feng B, Clinger OD, St Leger AJ, Liu X, Ghosh A, Dermond CD, Lathrop KL, Tochtrop GP, Picaud S, Chen Y. Non-retinoid chaperones improve rhodopsin homeostasis in a mouse model of retinitis pigmentosa. JCI Insight 2022; 7:153717. [PMID: 35472194 PMCID: PMC9220944 DOI: 10.1172/jci.insight.153717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Rhodopsin-associated (RHO-associated) retinitis pigmentosa (RP) is a progressive retinal disease that currently has no cure. RHO protein misfolding leads to disturbed proteostasis and the death of rod photoreceptors, resulting in decreased vision. We previously identified nonretinoid chaperones of RHO, including YC-001 and F5257-0462, by small-molecule high-throughput screening. Here, we profile the chaperone activities of these molecules toward the cell-surface level of 27 RP-causing human RHO mutants in NIH3T3 cells. Furthermore, using retinal explant culture, we show that YC-001 improves retinal proteostasis by supporting RHO homeostasis in RhoP23H/+ mouse retinae, which results in thicker outer nuclear layers (ONL), indicating delayed photoreceptor degeneration. Interestingly, YC-001 ameliorated retinal immune responses and reduced the number of microglia/macrophages in the RhoP23H/+ retinal explants. Similarly, F5257-0462 also protects photoreceptors in RhoP23H/+ retinal explants. In vivo, intravitreal injection of YC-001 or F5257-0462 microparticles in PBS shows that F5257-0462 has a higher efficacy in preserving photoreceptor function and delaying photoreceptor death in RhoP23H/+ mice. Collectively, we provide proof of principle that nonretinoid chaperones are promising drug candidates in treating RHO-associated RP.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Yibo Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Bing Feng
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Owen D Clinger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Anthony J St Leger
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Xujie Liu
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Archisha Ghosh
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Chase D Dermond
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Kira L Lathrop
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, United States of America
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, United States of America
| |
Collapse
|
45
|
Jiang M, Huang Y, Hu L, Wu H, Liu Y, Ni K, Zhang X, Sun Y, Gu X. The transcription factor CCAAT/enhancer-binding protein β in spinal microglia contributes to pre-operative stress-induced prolongation of postsurgical pain. Mol Pain 2022; 18:17448069221099360. [PMID: 35451875 PMCID: PMC9257637 DOI: 10.1177/17448069221099360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Prolongation of postsurgical pain caused by pre-operative stress is a clinically significant problem, although the mechanisms are not fully understood. Stress can promote the pro-inflammatory activation of microglia, and the transcription factor CCAAT/enhancer-binding protein (C/EBP) β regulates pro-inflammatory gene expression in microglia. Therefore, we speculated that C/EBPβ in spinal microglia may have critical roles in the development of chronic postsurgical pain. Accordingly, in this study, we used a single prolonged stress (SPS) procedure and plantar incisions to evaluate the roles of C/EBPβ in postsurgical pain. Our experiments showed that SPS exposure prolonged mechanical allodynia, increased the expression of C/EBPβ and pro-inflammatory cytokines, and potentiated the activation of spinal microglia. Subsequently, microinjection of C/EBPβ siRNA attenuated the duration of SPS-prolonged postoperative mechanical allodynia and inhibited microglial activation in the spinal cord. Conversely, mimicking this increase in C/EBPβ promoted microglial activation via pretreatment with a pre-injection of AAV5-C/EBPβ, leading to prolongation of postsurgical pain. Overall, these results suggested that spinal microglia may play key roles in prolongation of postsurgical pain induced by pre-operative stress and that C/EBPβ may be a potential target for disease treatment.
Collapse
Affiliation(s)
- Ming Jiang
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yulin Huang
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Lijun Hu
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Hao Wu
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yue Liu
- Department of Anesthesiology66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Kun Ni
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Xiaokun Zhang
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Yu'e Sun
- 66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| | - Xiaoping Gu
- Anesthesiology, Affiliated Drum Tower Hospital of Nanjing University Medical School66506Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital
| |
Collapse
|
46
|
Zhou HJ, Wang LQ, Zhan RY, Zheng XJ, Zheng JS. lncRNA MEG3 restrained the M1 polarization of microglia in acute spinal cord injury through the HuR/A20/NF-κB axis. Brain Pathol 2022; 32:e13070. [PMID: 35338543 PMCID: PMC9425005 DOI: 10.1111/bpa.13070] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/31/2021] [Accepted: 03/09/2022] [Indexed: 11/28/2022] Open
Abstract
The M1 polarization of microglia and neuroinflammation restrict the treatment of acute spinal cord injury (ASCI), and long non‐coding ribonucleic acid (lncRNA) maternally expressed gene 3 (MEG3) expression is lessened in ASCI. However, the function and mechanism of lncRNA MEG3 in the M1 polarization of microglia and neuroinflammation in ASCI are unclear. The expressions of lncRNA MEG3 in ASCI mouse spinal cord tissues and lipopolysaccharide (LPS)‐treated primary microglia and BV2 cells were quantified through a quantitative real‐time polymerase chain reaction. In‐vitro assays were conducted to explore the function of lncRNA MEG3 in the M1 polarization of microglia and neuroinflammation in ASCI. RNA degradation, RNA immunoprecipitation, RNA pull‐down, cycloheximide‐chase, and ubiquitination analyses were carried out to probe into the mechanism of lncRNA MEG3 in the M1 polarization of microglia and neuroinflammation in ASCI. The lncRNA MEG3 expression was lessened in the ASCI mouse spinal cord tissues and LPS‐treated primary microglia and BV2 cells, and the overexpression of lncRNA MEG3 restrained the M1 polarization of microglia and the neuroinflammation by regulating the NF‐κB signaling pathway. For the investigation of the potential mechanism of such, the overexpression of lncRNA MEG3 restrained the M1 polarization of microglia through the HuR/A20/NF‐κB axis and boosted the motor function recovery and neuroinflammation relief in the mice with SCI. The overexpression of lncRNA MEG3 restrained the M1 polarization of microglia through the HuR/A20/NF‐κB axis.
Collapse
Affiliation(s)
- Heng-Jun Zhou
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Li-Qing Wang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ren-Ya Zhan
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xiu-Jue Zheng
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jie-Sheng Zheng
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
47
|
Liu H, Wang LL, Xu QH, Wang J, Zhang YJ, Luo J, Liao AH. UHRF1 shapes both the trophoblast invasion and decidual macrophage differentiation in early pregnancy. FASEB J 2022; 36:e22247. [PMID: 35262949 DOI: 10.1096/fj.202101647rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/04/2022] [Accepted: 02/23/2022] [Indexed: 11/11/2022]
Abstract
Trophoblasts play critical roles in establishment and maintenance of a normal pregnancy. Their dysfunction in early pregnancy is closely related to pregnancy-related diseases, including recurrent pregnancy loss (RPL). Epigenetic modifications dynamically change during pregnancy; however, the role of the epigenetic modifier UHRF1 in trophoblast regulation remains unknown. This is the first study to show that UHRF1 expression was localized in the cytoplasm of cytotrophoblasts, syncytiotrophoblasts, and villi columns, and decreased in the villi of patients with RPL. The invasion and cell viability in a UHRF1 knockdown trophoblast cell line were significantly decreased. In addition, the mRNA expression profiles of Swan71 cells were partially altered by UHRF1 knockdown. The altered immune-related genes were screened out and the pro-inflammatory TH1-type chemokine/cytokines CXCL2 and IL-1β were identified as the most promising targets of UHRF1 in the trophoblasts, which were significantly increased in the UHRF1 knockdown Swan71 cells, villi, and serum from patients with RPL. The macrophages treated with the supernatants of UHRF1 knockdown Swan71 cells were polarized to the M1 phenotype and secreted high levels of pro-inflammatory cytokines, which might be driven by the activated MyD88/NF-κB signaling pathway and mediated by the increased expression of CXCR2 and IL-1R1 (CXCL2 and IL-1β receptors, respectively). In addition, the supernatants of UHRF1 knockdown Swan71 cells showed stronger chemotaxis to macrophages than those from the controls. Our findings highlight the previously unknown roles of UHRF1 as one of the key regulators on the trophoblasts and their cross-talk with local immune cells, and demonstrate a potential approach for RPL intervention.
Collapse
Affiliation(s)
- Hong Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Li-Ling Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Qian-Han Xu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yu-Jing Zhang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
48
|
Cai M, Lin W. The Function of NF-Kappa B During Epilepsy, a Potential Therapeutic Target. Front Neurosci 2022; 16:851394. [PMID: 35360161 PMCID: PMC8961383 DOI: 10.3389/fnins.2022.851394] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Abstract
The transcriptional regulator nuclear factor kappa B (NF-κB) modulates cellular biological activity by binding to promoter regions in the nucleus and transcribing various protein-coding genes. The NF-κB pathway plays a major role in the expressing genes related to inflammation, including chemokines, interleukins, and tumor necrosis factor. It also transcribes genes that can promote neuronal survival or apoptosis. Epilepsy is one of the most common brain disorders and it not only causes death worldwide but also affects the day-to-day life of affected individuals. While epilepsy has diverse treatment options, there remain patients who are not sensitive to the existing treatment methods. Recent studies have implicated the critical role of NF-κB in epilepsy. It is upregulated in neurons, glial cells, and endothelial cells, due to neuronal loss, glial cell proliferation, blood-brain barrier dysfunction, and hippocampal sclerosis through the glutamate and γ-aminobutyric acid imbalance, ion concentration changes, and other mechanisms. In this review, we summarize the functional changes caused by the upregulation of NF-κB in the central nervous system during different periods after seizures. This review is the first to deconvolute the complicated functions of NF-κB, and speculate that the regulation of NF-κB can be a safe and effective treatment strategy for epilepsy.
Collapse
|
49
|
Liu Y, Hu P, Zheng Z, Zhong D, Xie W, Tang Z, Pan B, Luo J, Zhang W, Wang X. Photoresponsive Vaccine-Like CAR-M System with High-Efficiency Central Immune Regulation for Inflammation-Related Depression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108525. [PMID: 34897839 DOI: 10.1002/adma.202108525] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/25/2021] [Indexed: 06/14/2023]
Abstract
Increasing evidence suggests that activation of microglia-induced neuroinflammation plays a crucial role in the pathophysiology of depression. Consequently, targeting the central nervous system to reduce neuroinflammation holds great promise for the treatment of depression. However, few drugs can enter the brain via a circulatory route through the blood-brain barrier (BBB) to reach the central nervous system efficiently, which limits the pharmacological treatment for neuropsychiatric diseases. Herein, a light-responsive system named UZPM, consisting of blue-emitting NaYF4 :Yb, Tm@zeolitic-imidazolate framework (UCNP@ZIF-8), photoacid (PA), and melatonin (MT) is developed to address the above issues. Meanwhile, UZPM is introduced into macrophages by functional liposomes fusion and modified with hydroxylamine groups on the cell surface. Aldehyde-modified cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is used as a chimeric antigen receptor (CAR) targeting group to modify the surface of macrophages by aldehyde/hydroxylamine condensation to precisely target central M1-type microglia (CAR-M-UZPM). Both in vitro and in vivo experiments demonstrate that the CAR-M-UZPM drug delivery system can efficiently penetrate the BBB, targeting centrally activated microglia, and thus, inhibiting the M1-type polarization of microglia, producing continuous vaccine-like anti-inflammatory effects that prevent the occurrence and development of inflammation-related depression.
Collapse
Affiliation(s)
- Yu Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, P. R. China
| | - Ping Hu
- Institute of Translational Medicine, School of Life Sciences, Nanchang University, Nanchang, 330088, P. R. China
| | - Zhiheng Zheng
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Da Zhong
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Weichang Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Zhibo Tang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| | - Bingxing Pan
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Jun Luo
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, 330006, P. R. China
| | - Wenhua Zhang
- Institute of Life Science, School of Life Sciences, Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, 330088, P. R. China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, P. R. China
| |
Collapse
|
50
|
Wu P, Zhou G, Wu X, Lv R, Yao J, Wen Q. P2X7 receptor induces microglia polarization to the M1 phenotype in cancer-induced bone pain rat models. Mol Pain 2022; 18:17448069211060962. [PMID: 35057643 PMCID: PMC8961217 DOI: 10.1177/17448069211060962] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background The transition from pro-inflammatory M1 phenotype to anti-inflammatory M2
phenotype presents a novel therapeutic strategy for chronic pain. Objective We investigated the role of microglia polarization in cancer-induced bone
pain (CIBP), as well as the role of the P2X7 receptor in modulating M1 to M2
polarization. Methods Walker-256 breast cancer cells were administered into tibias of female rats
to induce bone cancer–associated cancer. Results During bone cancer development, the P2X7 receptor and M1 microglia markers
were upregulated. In contrast, inhibition of the P2X7 receptor by BBG, a
blood-brain barrier-permeable P2X7R-specific antagonist, alleviated the pain
and promoted microglia polarization toward the M2 phenotype, while
suppressing the M1 phenotype in vivo and in
vitro. Conclusion P2X7 receptor-mediated spinal microglia polarization is involved in
alleviation of CIBP. Therefore, P2X7R is a potential option for CIBP
treatment.
Collapse
Affiliation(s)
- Ping Wu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guohua Zhou
- Anesthesiology Department, Dalian Medical University, Dalian, China
| | - Xiaoqi Wu
- Anesthesiology Department, Dalian Medical University, Dalian, China
| | - Run Lv
- Department of Anesthesiology, The first hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaqi Yao
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingping Wen
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|