1
|
Zhang Q, Yang L, Li C, Zhang Y, Li R, Jia F, Wang L, Ma X, Yao K, Tian H, Zhuo C. Exploring the potential antidepressant mechanisms of ibuprofen and celecoxib based on network pharmacology and molecular docking. J Affect Disord 2025; 377:136-147. [PMID: 39986574 DOI: 10.1016/j.jad.2025.02.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Evidence has shown that ibuprofen and celecoxib are effective in improving depressive symptoms, but their mechanisms of action are unclear. In this study, we aimed to determine the relationship between these two drugs and depressive disorder (DD) and elucidate potential mechanisms of action. METHODS Relevant targets for ibuprofen, celecoxib, and DD were obtained and screened from multiple online drug and disease public databases. A protein-protein interaction network was obtained. The Centiscape and CytoHubba plug-ins were applied to screen for core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed. Molecular docking was performed to predict the binding of ibuprofen and celecoxib to core targets. Examined the differences in core target protein expression between DD patients (DDs, n = 18) and healthy controls (HCs, n = 16) as a further experimental validation of the network pharmacology results. RESULTS In total, 220 potential targets for ibuprofen and 316 potential targets for celecoxib were identified and associated with DD. The antidepressant effects of both drugs involve many key targets in pathways such as "pathways in cancer" and "neuroactive ligand-receptor interaction," including ALB, BCL2, MAPK3, SRC, STAT3, EGFR, and PPARG. The binding affinity of ALB with ibuprofen is the strongest, and it is connected only by hydrophobic interactions. Celecoxib exhibits higher affinity at multiple targets such as SRC, EGFR, and PPARG, with stronger and more specific intermolecular interactions, including salt bridges and halogen bonds. Clinical trials have found that serum ALB expression in DDs is significantly lower than that in HCs (t = 6.653, p < 0.001), further confirming the potential role of ibuprofen in DD. CONCLUSIONS Ibuprofen and celecoxib primarily exert their antidepressant effects through targets and pathways related to inflammation, neural signaling, and cancer, with celecoxib showing a stronger potential antidepressant effect. The expression difference of the core target ALB between depression and healthy individuals further supports the potential effect of the drug on DD. Our findings propose new treatment strategies, support the link between inflammation and depression, and encourage reassessing existing medications for depression.
Collapse
Affiliation(s)
- Qiuyu Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lei Yang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chao Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ying Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ranli Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Feng Jia
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lina Wang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Xiaoyan Ma
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Kaifang Yao
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Hongjun Tian
- Animal Imaging Center (AIC) of Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China
| | - Chuanjun Zhuo
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| |
Collapse
|
2
|
Chen S, Li Z, Yang L, Xu Z, Liu A, He Q, Xiao F, Zhan J. Cannabinoid Receptor-2 Alleviates Sepsis-Induced Neuroinflammation by Modulating Microglia M1/M2 Subset Polarization Through Inhibiting Nogo-B Expression. Mol Neurobiol 2025:10.1007/s12035-025-04836-2. [PMID: 40102346 DOI: 10.1007/s12035-025-04836-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
Few studies have investigated how Nogo-B affects sepsis-associated encephalopathy (SAE). Cannabinoid receptor 2 (CB2R) plays a critical role in regulating M1/M2 polarization in microglia. This study aimed to explore the association between CB2R and Nogo-B by assessing changes in microglial polarization markers.C57BL/6 mice with SAE induced by cecal ligation and puncture (CLP) surgery were intraperitoneally injected with HU308 for 3 consecutive days at the same time after that, and changes in cognitive function were assessed. After Lipopolysaccharides (LPS) and Interleukin-4 (IL-4) were used to induce BV2 microglial cell models respectively, HU308 and AM630 were applied to assess changes in inflammatory factors, microglial polarization markers, and the expression levels of CB2R and Nogo-B in microglial cells. We established a stable Nogo-B overexpression cell line. ELISA, Western blot, and flow cytometry were utilized to verify whether Nogo-B is a crucial protein in controlling BV2 cell polarization by HU308. There was an increase in Nogo-B protein expression during SAE. HU308 treatment alleviated the cognitive impairment of the CLP mice and markedly decreased the level of Nogo-B in the hippocampus tissues. The efficacy of CB2R activation to promote microglia polarization from M1 to M2 was diminished in BV2 cells overexpressing Nogo-B, although its anti-inflammatory effect was not entirely reversed. Inhibiting the Nogo-B expression, which in turn encourages the conversion of BV2 microglia to M2, attenuates inflammatory responses, and promotes neuronal repair, could be a key mechanism whereby activation of CB2R ameliorates septic encephalopathy.
Collapse
Affiliation(s)
- Shuxian Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Zhen Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Liu Yang
- Department of Anesthesiology, Wuhan Asian Heart Hospital, 430022, Wuhan, Hubei, People's Republic of China
| | - Zujin Xu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Anpeng Liu
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Qianwen He
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 430071, Wuhan, Hubei, People's Republic of China
| | - Fei Xiao
- Department of Orthopedics, Wuhan Fourth Hospital, 473 Hanzheng Street, Qiaokou District, Wuhan, 430033, Hubei, People's Republic of China.
| | - Jia Zhan
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, 169, East-Lake Road, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
3
|
Lei X, Zhao D, Chen T, Li Q, Xue A, Hu Z, Jia F, Li X. Exploring the active components and potential mechanisms of Zhimu-Huangbai herb-pair in the treatment of depression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156365. [PMID: 39904199 DOI: 10.1016/j.phymed.2025.156365] [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: 09/01/2024] [Revised: 12/15/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The Zhimu-Huangbai herb-pair (ZB) is commonly used to treat depression. Previous research has verified that ZB is effective as an antidepressant. Nevertheless, its active components and potential mechanism still require further elucidation. PURPOSE This study aims to analyze the compounds of ZB penetrating into the brain using UPLC-MS and investigate the potential mechanisms of ZB in the treatment of depression through in vivo and in vitro experiments. METHODS The compounds of ZB that penetrate into the brain were identified using the UPLC-MS method. Network pharmacology analysis was employed to predict the therapeutic targets and mechanisms of the compounds of ZB in the brain for the treatment of depression. Subsequently, the molecular docking method was used to analyze the binding between active compounds and target proteins. Rat depression models induced by CUMS were used to investigate the impact of ZB on depression. Finally, the mechanism of ZB treatment for depression was investigated using the LPS-induced BV2 cell inflammation model. RESULTS A total of 17 compounds were identified in ZB that crossed the blood-brain barrier (BBB). The network pharmacological analysis showed that the anti-depressant mechanism of ZB is closely related to inflammatory cytokines, including TNF and IL-6. Furthermore, KEGG and PPI analyses demonstrated that ZB regulates the microglia M1/M2 phenotypic polarization by modulating inflammation-related pathways. ZB was found to improve depression-like behavior in vivo. The molecular docking indicated that the compounds in ZB that penetrate into the brain have a strong binding ability to RELA and PPAR-γ. ZB inhibited the expression of p-p65 and increased the expression of PPAR-γ in the mPFC. By rebalancing the ratio of pro-inflammatory/anti-inflammatory cytokines, ZB was able to reduce neuroinflammation in the mPFC and hippocampus regions. The immunofluorescence results showed that ZB-containing serum reduced M1 polarization induced by LPS in BV2 cells. CONCLUSION This study reveals that ZB effectively alleviates depression by regulating the M1/M2 phenotypic polarization of microglial cells. The mechanism may be that the active compounds of ZB reduce M1 phenotypic polarization by inhibiting P65 and increase M2 phenotypic polarization by promoting PPARγ.
Collapse
Affiliation(s)
- Xia Lei
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Deping Zhao
- College of Pharmacy, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Tongtong Chen
- College of Pharmacy, Heilongjiang University of Chinese Medicine, 150040 Harbin, Heilongjiang, China
| | - Qing Li
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Ao Xue
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Zhuoyi Hu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Fan Jia
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, 214071 Wuxi, Jiangsu, China
| | - Xiaoliang Li
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, 571199, China.
| |
Collapse
|
4
|
Chen Z, Ruan F, Wu D, Yu X, Jiang Y, Bao W, Wen H, Hu J, Bi H, Chen L, Le K. Quercetin alleviates neonatal hypoxic-ischaemic brain injury by rebalancing microglial M1/M2 polarization through silent information regulator 1/ high mobility group box-1 signalling. Inflammopharmacology 2025; 33:865-883. [PMID: 39565473 DOI: 10.1007/s10787-024-01599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/02/2024] [Indexed: 11/21/2024]
Abstract
Neonatal hypoxic-ischaemic encephalopathy (HIE) remains one of the major causes of neonatal death and long-term neurological disability. Due to its complex pathogenesis, there are still many challenges in its treatment. In our previous studies, we found that quercetin can alleviate neurological dysfunction after hypoxic-ischaemic brain injury (HIBI) in neonatal mice. As demonstrated through in vitro experiments, quercetin can inhibit the activation of the TLR4/MyD88/NF-κB signalling pathway and the inflammatory response in the microglial cell line BV2 after oxygen-glucose deprivation. However, the in-depth mechanism still needs to be further elucidated. In the present study, 7 day-old neonatal ICR mice or BV2 cells were treated with quercetin with or without the SIRT1 inhibitor EX527 via neurobehavioural, histopathological and molecular methods. In vivo experiments have shown that quercetin can significantly improve the performance of HI mice in behavioural tests, such as the Morris water maze, rotarod test and pole climbing test, and reduce HI insult-induced structural brain damage, cell apoptosis and hippocampal neuron loss. Quercetin also inhibited the immunofluorescence intensity of the microglial M1 marker CD16 + 32 and significantly downregulated the expression of the M1-related proteins iNOS, IL-1β and TNF-α. Moreover, quercetin increased the immunofluorescence intensity of the microglial M2 marker CD206 and significantly increased the expression of the M2-related proteins Arg-1 and IL-10. In addition, quercetin limits the nucleocytoplasmic translocation and release of microglial HMGB1 and further suppresses the activation of the downstream TLR4/MyD88/NF-κB signalling pathway. The above effects of quercetin are partially weakened by pretreatment with EX527. Similar results were found in in vitro experiments, and the mechanism further revealed that the rebalancing effect of quercetin on microglial polarization is achieved through the SIRT1-mediated reduction in HMGB1 acetylation levels. This study provides new and complementary insights into the neuroprotective effects of quercetin and a new direction for the treatment of neonatal HIE.
Collapse
Affiliation(s)
- Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Haicheng Wen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Jing Hu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Haidi Bi
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Liping Chen
- Department of Neonatology, Jiangxi Children's Hospital, No.122 Yangming Road, Nanchang, 330006, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China.
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hong Kong S.A.R., Hong Kong, China.
| |
Collapse
|
5
|
Yan L, Xu K, Liu C, Yu F, Guo J, Hou L, Feng Y, Yang M, Gong Q, Qin D, Qin M, Wang Y, Su H, Lu Y. Polymer-Formulated Nerve Growth Factor Shows Effective Therapeutic Efficacy for Cerebral Microinfarcts. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412843. [PMID: 39601176 DOI: 10.1002/adma.202412843] [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: 08/28/2024] [Revised: 10/04/2024] [Indexed: 11/29/2024]
Abstract
Cerebral microinfarcts represent the most prevalent form of ischemic brain injury in the elderly, particularly among those suffering from dementia, Alzheimer's disease, and vascular risk factors. Despite their commonality, effective treatments have remained elusive. Herein, a novel treatment utilizing a polymer-formulated nerve growth factor capable of crossing the blood-brain barrier is reported, which effectively reduced oxidative stress and neuronal apoptosis, reshaped microglia polarization at infarct sites, and decreased the overall microinfarct burden, leading to notable improvements in behavioral and cognitive functions in a mouse model. This work provides a promising new avenue for the treatment of cerebral microinfarcts and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Kunyao Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Jimin Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, 102206, China
| | - Yicheng Feng
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Mo Yang
- Department of Neurology, Beijing Tiantan Hospital, Beijing, 100071, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
| | - Dajiang Qin
- Center for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, 999077, China
| | - Meng Qin
- Mental Health Center and Center for Preclinical Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Beijing, 100071, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Yunfeng Lu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
6
|
Lin YE, Lin HP, Lu KH, Huang YJ, Panyod S, Liu WT, Lu YS, Chen MH, Sheen LY. Cordyceps militaris and Armillaria mellea formula alleviates depressive behaviors via microglia regulation in an unpredictable chronic mild stress animal model. J Tradit Complement Med 2025; 15:24-35. [PMID: 39807265 PMCID: PMC11725130 DOI: 10.1016/j.jtcme.2024.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 01/16/2025] Open
Abstract
Background and aim Cordyceps militaris (CM) and Armillaria mellea (AM) are medicinal mushrooms with potential applications in the treatment of mood disorders, including depression and anxiety. While research suggests that both CM and AM possess anti-inflammatory properties and hold potential for treating depression when administered separately, there is limited knowledge about their efficacy when combined in a formula, as well as the underlying mechanism involving the modulation of microglia. Experimental procedure Rats received oral administrations of the low-dose formulation, medium-dose formulation, and high-dose formulation over 28 consecutive days as part of the UCMS protocols. The concentrations of serotonin, dopamine, and the corresponding metabolites in the rat prefrontal cortex and hippocampus were assessed. Blood samples were collected to examine corticosterone levels, and the brains were dissected for evaluating activated microglia morphologies and associated pro- and anti-inflammatory signaling pathways. Results and conclusion The CM-AM formula effectively averted abnormal behaviors triggered by UCMS, such as anhedonia and hypoactivity, and decreased the turnover rate of monoamines in both the prefrontal cortex and hippocampus. The formula mitigated the increase in serum corticosterone levels induced by chronic stress. Furthermore, the formula alleviated stress-induced microglia activation in the hippocampus, achieving this by down-regulating hyperactivated pro-inflammatory proteins and up-regulating hypoactivated anti-inflammatory proteins in the hippocampus. The antidepressant-like effects potentially stemming from the regulation of neurotransmitters and immunomodulation, likely by restoring the balance of M1 and M2 microglia fractions in the hippocampus. Consequently, the CM-AM formula could be explored as a prospective complementary and alternative therapy for depression.
Collapse
Affiliation(s)
- Yu-En Lin
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Hui-Ping Lin
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Kuan-Hung Lu
- Institute of Food Safety and Health, National Taiwan University, Taipei, Taiwan
- Institute of Environmental and Occupational Health Sciences, National Taiwan University, Taipei, Taiwan
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Suraphan Panyod
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Wei-Ting Liu
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Yun-Sheng Lu
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Mei-Hsing Chen
- Taiwan Agricultural Research Institute, Council of Agricultural, Taichung, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
7
|
Jensen P, Ozenne B, Meden P, Feng L, Thomsen G, Knudsen L, Steglich‐Arnholm H, Møller K, Thomsen C, Svarer C, Beliveau V, Mikkelsen J, Knudsen G, H Pinborg L. Poststroke Translocator Protein Expression Dynamics and Correlations to Chronic Infarction: A [123I]-CLINDE-SPECT Study. J Neuroimaging 2025; 35:e70002. [PMID: 39803801 PMCID: PMC11726615 DOI: 10.1111/jon.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND PURPOSE This study aims to investigate the longitudinal changes in translocator protein (TSPO) following stroke in different brain regions and potential associations with chronic brain infarction. METHODS Twelve patients underwent SPECT using the TSPO tracer 6-Chloro-2-(4'-123I-Iodophenyl)-3-(N,N-Diethyl)-Imidazo[1,2-a]Pyridine-3-Acetamide, as well as structural MRI, at 10, 41, and 128 days (median) after ischemic infarction in the middle cerebral artery. TSPO expression was measured in lesional (MRI lesion and SPECT lesion), connected (pons and ipsilesional thalamus), and nonconnected (ipsilesional cerebellum and contralesional occipital cortex) regions. Correlations were explored between the volume of chronic infarction and TSPO expression in nonconnected regions of interest (ROIs) at 128 days RESULTS: Throughout the study period, TSPO levels decreased by 24%-33% in lesional ROIs, while levels increased in connected ROIs by 35%-69% and in nonconnected ROIs by 53%-77%. At 128 days poststroke, TSPO expression in ipsilesional cerebellum positively correlated with chronic infarction volume (p = 0.002, r2 = 0.72). CONCLUSIONS This study expands the current knowledge of spatial and temporal TSPO expression in humans by quantifying TSPO changes in lesional, connected, and nonconnected brain regions at three time points after cerebral infarction as well as correlating late-stage TSPO upregulation and chronic infarction volume.
Collapse
Affiliation(s)
- Per Jensen
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
- Epilepsy ClinicRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Brice Ozenne
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
- Department of Public Health, Section of BiostatisticsUniversity of CopenhagenCopenhagenDenmark
| | - Per Meden
- Department of NeurologyBispebjerg Hospital, University of CopenhagenCopenhagenDenmark
| | - Ling Feng
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Gerda Thomsen
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Lars Knudsen
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
| | | | - Kirsten Møller
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of NeuroanaesthesiologyRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Carsten Thomsen
- Department of RadiologyRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Claus Svarer
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Vincent Beliveau
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
| | - Jens Mikkelsen
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
- Faculty of Health and MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Gitte Knudsen
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
- Faculty of Health and MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Lars H Pinborg
- Neurobiology Research UnitRigshospitalet, University of CopenhagenCopenhagenDenmark
- Epilepsy ClinicRigshospitalet, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
8
|
Li H, Yu W, Zheng X, Zhu Z. TREM1-Microglia crosstalk: Neurocognitive disorders. Brain Res Bull 2025; 220:111162. [PMID: 39645047 DOI: 10.1016/j.brainresbull.2024.111162] [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: 08/26/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Neurocognitive Disorders (NCDs) primarily affect cognitive functions, including learning, memory, perception, and problem-solving. They predominantly arise as pathological sequelae of central nervous system (CNS) disorders. Emerging evidence suggests that microglial inflammatory activation within the hippocampus underlies the pathogenesis of cognitive impairment. Triggering receptor expressed on myeloid cells 1 (TREM1), a pattern-recognition receptor on microglia, becomes upregulated in response to injury and synergistically amplifies inflammatory responses mediated by other pattern-recognition receptors, leading to uncontrolled inflammation. While TREM1 is lowly expressed in the resting state, its upregulation upon exposure to injurious inflammatory stimuli promotes microglial activation and contributes to the development of NCDs. Consequently, TREM1 may serve as a critical receptor in microglia-mediated inflammation. This article reviews the current understanding of TREM1 and its role in NCDs pathogenesis.
Collapse
Affiliation(s)
- Huashan Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China; Department of Anesthesiology, Zunyi Maternal And Child Health Care Hospital, Zunyi 563000, China.
| | - Wanqiu Yu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China
| | - Xue Zheng
- Department of Anesthesiology, Zunyi Maternal And Child Health Care Hospital, Zunyi 563000, China
| | - Zhaoqiong Zhu
- Early Clinical Research Ward, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, China.
| |
Collapse
|
9
|
Tao Y, Sun Y, Jiang X, Tao J, Zhang Y. The Role of Alpha-7 Nicotinic Acetylcholine Receptors in Pain: Potential Therapeutic Implications. Curr Neuropharmacol 2025; 23:129-144. [PMID: 38808717 PMCID: PMC11793049 DOI: 10.2174/1570159x22666240528161117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/21/2024] [Accepted: 01/21/2024] [Indexed: 05/30/2024] Open
Abstract
Chronic pain represents a prevalent and costly medical challenge globally. Nicotinic acetylcholine receptors (nAChRs), one type of ligand-gated ion channels found extensively in both the central and peripheral nervous systems, have emerged as promising therapeutic targets for chronic pain. Although there are currently no FDA-approved analgesics specifically targeting nAChRs, accumulating preclinical and clinical evidence suggest that selective ligands for alpha 7 (α7) nAChRs show potential for treating chronic pain, boasting a reduced incidence of side effects compared with other nicotinic receptor types. The recent structural resolution of human α7 nAChRs has confirmed their negative association with heightened pain, providing a valuable foundation for the development of targeted medications. This review presents a comprehensive overview, encompassing insights into the roles of α7 nAChRs derived from structural and functional studies, recent advancements in pharmacology, and investigations into their involvement in the pathophysiology of chronic pain. Moreover, the review addresses the variability in analgesic effects based on the type of receptor agonist and highlights the current research limitations. As such, this review offers potential therapeutic approaches for the development of innovative strategies for chronic pain management.
Collapse
Affiliation(s)
- Yu Tao
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P.R. China
- Department of Physiology and Neurobiology, Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, P.R. China
| | - Yufang Sun
- Department of Physiology and Neurobiology, Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, P.R. China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology, Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, P.R. China
| | - Jin Tao
- Department of Physiology and Neurobiology, Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, P.R. China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, P.R. China
| | - Yuan Zhang
- Clinical Research Center of Neurological Disease, Department of Geriatrics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, P.R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, P.R. China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, P.R. China
| |
Collapse
|
10
|
Zhou W, Chang Y, Xiao Q, Deng Z, Zhang L, Yuan Z, Du Z. Structural optimization of naturally derived Ar-turmerone, as novel neuroinflammation suppressors effective in an Alzheimer mouse model. Bioorg Med Chem 2025; 117:118014. [PMID: 39602866 DOI: 10.1016/j.bmc.2024.118014] [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: 08/27/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Microglia-mediated neuroinflammation plays a pivotal role in neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. The modulation of chronic and sustained inflammatory processes in the brain with small molecules presents a promising therapeutic strategy for these devastating conditions. Aromatic turmerone (ar-turmerone, ART), an active constituent of turmeric essential oil derived from the edible plant Curcuma longa, has shown substantial potential in mitigating neuroinflammatory responses and associated cognitive deficits. Building on our previous work, we sought to discover more potent neuroinflammation suppressors by designing and synthesizing a series of ar-turmerone derivatives to investigate their structure-activity relationships. Microglia-based cellular evaluations revealed that naphthyl-substituted (7c) and N-substituted amides (7a) demonstrated the most pronounced inhibitory effects against NO, TNF-α, and IL-1β release in vitro. Furthermore, in a lipopolysaccharide (LPS)-induced neuroinflammation model of Alzheimer's disease in mice, these two compounds significantly reduced proinflammatory cytokine release, protected neurons from damage, and ameliorated memory impairments and cognitive deficits in Morris water maze tests. This structural optimization of ar-turmerone yielded highly potent anti-neuroinflammatory compounds, which may serve as promising agents for the treatment of neuroinflammation-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Wei Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China.
| | - Yuanyuan Chang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China
| | - Qingwei Xiao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China
| | - Zhujie Deng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China
| | - Lanyue Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China.
| | - Zhiyun Du
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
11
|
Darmanto AG, Yen TL, Jan JS, Linh TTD, Taliyan R, Yang CH, Sheu JR. Beyond metabolic messengers: Bile acids and TGR5 as pharmacotherapeutic intervention for psychiatric disorders. Pharmacol Res 2025; 211:107564. [PMID: 39733841 DOI: 10.1016/j.phrs.2024.107564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Psychiatric disorders pose a significant global health challenge, exacerbated by the COVID-19 pandemic and insufficiently addressed by the current treatments. This review explores the emerging role of bile acids and the TGR5 receptor in the pathophysiology of psychiatric conditions, emphasizing their signaling within the gut-brain axis. We detail the synthesis and systemic functions of bile acids, their transformation by gut microbiota, and their impact across various neuropsychiatric disorders, including major depressive disorder, general anxiety disorder, schizophrenia, autism spectrum disorder, and bipolar disorder. The review highlights how dysbiosis and altered bile acid metabolism contribute to the development and exacerbation of these neuropsychiatric disorders through mechanisms involving inflammation, oxidative stress, and neurotransmitter dysregulation. Importantly, we detail both pharmacological and non-pharmacological interventions that modulate TGR5 signaling, offering potential breakthroughs in treatment strategies. These include dietary adjustments to enhance beneficial bile acids production and the use of specific TGR5 agonists that have shown promise in preclinical and clinical settings for their regulatory effects on critical pathways such as cAMP-PKA, NRF2-mediated antioxidant responses, and neuroinflammation. By integrating findings from the dynamics of gut microbiota, bile acids metabolism, and TGR5 receptor related signaling events, this review underscores cutting-edge therapeutic approaches poised to revolutionize the management and treatment of psychiatric disorders.
Collapse
Affiliation(s)
- Arief Gunawan Darmanto
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; School of Medicine, Universitas Ciputra, Surabaya 60219, Indonesia
| | - Ting-Lin Yen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Department of Medical Research, Cathay General Hospital, Taipei 22174, Taiwan, ROC
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC
| | - Tran Thanh Duy Linh
- Family Medicine Training Center, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Viet Nam
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC.
| | - Joen-Rong Sheu
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan, ROC; Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan, ROC; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan, ROC.
| |
Collapse
|
12
|
Yang Y, Wu J, Jia L, Feng S, Qi Z, Yu H, Wu Y, Wang S. Berberine modulates microglial polarization by activating TYROBP in Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156237. [PMID: 39566407 DOI: 10.1016/j.phymed.2024.156237] [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: 07/30/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Characterized by β-amyloid (Aβ) plaques, neurofibrillary tangles, and aberrant neuroinflammation in the brain, Alzheimer's disease (AD) is the most common neurodegenerative disease. Microglial polarization is a subtle mechanism which maintains immunological homeostasis and has emerged as a putative therapeutic to combat AD. Berberine (BBR) is a natural alkaloid compound with multiple pharmacological effects, and has shown considerable therapeutic potential against inflammatory disorders. However, BBR functions and underlying mechanisms in neuroinflammation remain unclear. PURPOSE To examine BBR pharmacological effects and mechanisms in neuroinflammation with a view to treating AD. METHODS BBR effects on cognitive performance in 5 × FAD mice were assessed using open field, Y-maze, and Morris Water Maze (MWM) tests. Neuroinflammation-related markers and Aβ pathology were examined in brain sections from mice. Transcriptomic analyses of hippocampus tissues were also conducted. Microglial BV2 cells were also used to verify potential BBR mechanisms in neuroinflammation and microglial polarization. RESULTS BBR improved cognitive performance, reduced amyloid pathology, and alleviated aberrant neuroinflammation in an AD mouse model. BBR induced microglial polarization to an M2-like phenotype, which was manifested by lowered and elevated proinflammatory and anti-inflammatory cytokine production, respectively, improved microglial uptake and Aβ clearance. Mechanistically, BBR directly interacted with TYROBP and promoted its activation by stabilizing TYROBP oligomerization. TYROBP knockdown aggravated M1-like polarization and pro-inflammatory gene expression in microglial cells in the presence of lipopolysaccharide (LPS)+Aβ, while blocked microglial M2-like polarization benefited from BBR administration. CONCLUSIONS BBR modulated neuroinflammation by regulating microglial polarization via TYROBP activation. Our study provided new insight into BBR pharmacological actions in regulating microglial homeostasis and combating AD.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Jiwen Wu
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Luping Jia
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Shicheng Feng
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Zihan Qi
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Hao Yu
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China
| | - Yili Wu
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Key Laboratory of Basic and Translational Research for Mental Disorders, Zhejiang Provincial Clinical Research Center for Mental Health, School of Mental Health, Institute of Aging, Wenzhou Medical University, Wenzhou, 325000, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, 325000, China.
| | - Shuai Wang
- Shandong Key Laboratory of Psychiatric and Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, 272013, Shandong, China; Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, 272013, Shandong, China.
| |
Collapse
|
13
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Effects and mechanisms of APP and its cleavage product Aβ in the comorbidity of sarcopenia and Alzheimer's disease. Front Aging Neurosci 2024; 16:1482947. [PMID: 39654807 PMCID: PMC11625754 DOI: 10.3389/fnagi.2024.1482947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Sarcopenia and AD are both classic degenerative diseases, and there is growing epidemiological evidence of their comorbidity with aging; however, the mechanisms underlying the biology of their commonality have not yet been thoroughly investigated. APP is a membrane protein that is expressed in tissues and is expressed not only in the nervous system but also in the NMJ and muscle. Deposition of its proteolytic cleavage product, Aβ, has been described as a central component of AD pathogenesis. Recent studies have shown that excessive accumulation and aberrant expression of APP in muscle lead to pathological muscle lesions, but the pathogenic mechanism by which APP and its proteolytic cleavage products act in skeletal muscle is less well understood. By summarizing and analyzing the literature concerning the role, pathogenicity and pathological mechanisms of APP and its cleavage products in the nervous system and muscles, we aimed to explore the intrinsic pathological mechanisms of myocerebral comorbidities and to provide new perspectives and theoretical foundations for the prevention and treatment of AD and sarcopenia comorbidities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| |
Collapse
|
14
|
Zhou H, Yang RK, Li Q, Li Z, Wang YC, Li SY, Miao Y, Sun XH, Wang Z. MicroRNA-146a-5p protects retinal ganglion cells through reducing neuroinflammation in experimental glaucoma. Glia 2024; 72:2115-2141. [PMID: 39041109 DOI: 10.1002/glia.24600] [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: 01/30/2024] [Revised: 06/27/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Neuroinflammation plays important roles in retinal ganglion cell (RGC) degeneration in glaucoma. MicroRNA-146 (miR-146) has been shown to regulate inflammatory response in neurodegenerative diseases. In this study, whether and how miR-146 could affect RGC injury in chronic ocular hypertension (COH) experimental glaucoma were investigated. We showed that in the members of miR-146 family only miR-146a-5p expression was upregulated in COH retinas. The upregulation of miR-146a-5p was observed in the activated microglia and Müller cells both in primary cultured conditions and in COH retinas, but mainly occurred in microglia. Overexpression of miR-146a-5p in COH retinas reduced the levels pro-inflammatory cytokines and upregulated the levels of anti-inflammatory cytokines, which were further confirmed in the activated primary cultured microglia. Transfection of miR-146a-5p mimic increased the percentage of anti-inflammatory phenotype in the activated BV2 microglia, while transfection of miR-146a-5p inhibitor resulted in the opposite effects. Transfection of miR-146a-5p mimic/agomir inhibited the levels of interleukin-1 receptor associated kinase (IRAK1) and TNF receptor associated factor 6 (TRAF6) and phosphorylated NF-κB subunit p65. Dual luciferase reporter gene assay confirmed that miR-146a-5p could directly target IRAK1 and TRAF6. Moreover, downregulation of IRAK1 and TRAF6 by siRNA techniques or blocking NF-κB by SN50 in cultured microglia reversed the miR-146a-5p inhibitor-induced changes of inflammatory cytokines. In COH retinas, overexpression of miR-146a-5p reduced RGC apoptosis, increased RGC survival, and partially rescued the amplitudes of photopic negative response. Our results demonstrate that overexpression of miR-146a-5p attenuates RGC injury in glaucoma by reducing neuroinflammation through downregulating IRAK1/TRAF6/NF-κB signaling pathway in microglia.
Collapse
Affiliation(s)
- Han Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Rui-Kang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Qian Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhen Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yong-Chen Wang
- Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Shu-Ying Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xing-Huai Sun
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, NHC Key Laboratory of Myopia, Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Yin Y, Ju T, Zeng D, Duan F, Zhu Y, Liu J, Li Y, Lu W. "Inflamed" depression: A review of the interactions between depression and inflammation and current anti-inflammatory strategies for depression. Pharmacol Res 2024; 207:107322. [PMID: 39038630 DOI: 10.1016/j.phrs.2024.107322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Depression is a common mental disorder, the effective treatment of which remains a challenging issue worldwide. The clinical pathogenesis of depression has been deeply explored, leading to the formulation of various pathogenic hypotheses. Among these, the monoamine neurotransmitter hypothesis holds a prominent position, yet it has significant limitations as more than one-third of patients do not respond to conventional treatments targeting monoamine transmission disturbances. Over the past few decades, a growing body of research has highlighted the link between inflammation and depression as a potential key factor in the pathophysiology of depression. In this review, we first summarize the relationship between inflammation and depression, with a focus on the pathophysiological changes mediated by inflammation in depression. The mechanisms linking inflammation to depression as well as multiple anti-inflammatory strategies are also discussed, and their efficacy and safety are assessed. This review broadens the perspective on specific aspects of using anti-inflammatory strategies for treating depression, laying the groundwork for advancing precision medicine for individuals suffering from "inflamed" depression.
Collapse
Affiliation(s)
- Yishu Yin
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Ting Ju
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Deyong Zeng
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Fangyuan Duan
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Yuanbing Zhu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China
| | - Junlian Liu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yongzhi Li
- China Astronaut Research and Training Center, Beijing 100094, China.
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin 150001, China.
| |
Collapse
|
16
|
Qiu R, Cai Y, Su Y, Fan K, Sun Z, Zhang Y. Emerging insights into Lipocalin-2: Unraveling its role in Parkinson's Disease. Biomed Pharmacother 2024; 177:116947. [PMID: 38901198 DOI: 10.1016/j.biopha.2024.116947] [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: 03/03/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Parkinson's disease (PD) ranks as the second most prevalent neurodegenerative disorder globally, marked by a complex pathogenesis. Lipocalin-2 (LCN2) emerges as a crucial factor during the progression of PD. Belonging to the lipocalin family, LCN2 is integral to several biological functions, including glial cell activation, iron homeostasis regulation, immune response, inflammatory reactions, and oxidative stress mitigation. Substantial research has highlighted marked increases in LCN2 expression within the substantia nigra (SN), cerebrospinal fluid (CSF), and blood of individuals with PD. This review focuses on the pathological roles of LCN2 in neuroinflammation, aging, neuronal damage, and iron dysregulation in PD. It aims to explore the underlying mechanisms of LCN2 in the disease and potential therapeutic targets that could inform future treatment strategies.
Collapse
Affiliation(s)
- Ruqing Qiu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yunjia Cai
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yana Su
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangli Fan
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
17
|
Yu Z, Shi H, Zhang J, Ma C, He C, Yang F, Zhao L. ROLE OF MICROGLIA IN SEPSIS-ASSOCIATED ENCEPHALOPATHY PATHOGENESIS: AN UPDATE. Shock 2024; 61:498-508. [PMID: 38150368 DOI: 10.1097/shk.0000000000002296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
ABSTRACT Sepsis-associated encephalopathy (SAE) is a serious complication of sepsis, which is characterized by cognitive dysfunction, a poor prognosis, and high incidences of morbidity and mortality. Substantial levels of systemic inflammatory factors induce neuroinflammatory responses during sepsis, ultimately disrupting the central nervous system's (CNS) homeostasis. This disruption results in brain dysfunction through various underlying mechanisms, contributing further to SAE's development. Microglia, the most important macrophage in the CNS, can induce neuroinflammatory responses, brain tissue injury, and neuronal dysregulation, resulting in brain dysfunction. They serve an important regulatory role in CNS homeostasis and can be activated through multiple pathways. Consequently, activated microglia are involved in several pathogenic mechanisms related to SAE and play a crucial role in its development. This article discusses the role of microglia in neuroinflammation, dysfunction of neurotransmitters, disruption of the blood-brain barrier, abnormal control of cerebral blood flow, mitochondrial dysfunction, and reduction in the number of good bacteria in the gut as main pathogenic mechanisms of SAE and focuses on studies targeting microglia to ameliorate SAE to provide a theoretical basis for targeted microglial therapy for SAE.
Collapse
Affiliation(s)
| | - Hui Shi
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Jingjing Zhang
- Department of Central Laboratory, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Chunhan Ma
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Chen He
- Chifeng Clinical Medical College of Inner Mongolia Medical University, Hohhot, China
| | - Fei Yang
- Department of Critical Care Medicine, Chifeng Municipal Hospital, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, China
| | - Lina Zhao
- Department of Critical Care Medicine, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
18
|
Charoensaensuk V, Huang BR, Huang ST, Lin C, Xie SY, Chen CW, Chen YC, Cheng HT, Liu YS, Lai SW, Shen CK, Lin HJ, Yang LY, Lu DY. LPS priming-induced immune tolerance mitigates LPS-stimulated microglial activation and social avoidance behaviors in mice. J Pharmacol Sci 2024; 154:225-235. [PMID: 38485340 DOI: 10.1016/j.jphs.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024] Open
Abstract
In this study, we investigated the regulatory mechanisms underlying the effects of LPS tolerance on the inflammatory homeostasis of immune cells. LPS priming-induced immune tolerance downregulated cyclooxygenase-2, and lowered the production of prostaglandin-E2 in microglial cells. In addition, LPS tolerance downregulated the expression of suppressor of cytokine signaling 3, and inducible nitric oxide synthase/nitric oxide; suppressed the LPS-mediated induction of tumor necrosis factor-α, interleukin (IL)-6, and IL-1; and reduced reactive oxygen species production in microglial cells. LPS stimulation increased the levels of the adaptive response-related proteins heme oxygenase-1 and superoxide dismutase 2, and the levels of heme oxygenase-1 (HO-1) enhanced after LPS priming. Systemic administration of low-dose LPS (0.5 mg/kg) to mice for 4 consecutive days attenuated high-dose LPS (5 mg/kg)-induced inflammatory response, microglial activation, and proinflammatory cytokine expression. Moreover, repeated exposure to low-dose LPS suppressed the recruitment of peripheral monocytes or macrophages to brain regions and downregulated the expression of proinflammatory cytokines. Notably, LPS-induced social avoidance behaviors in mice were mitigated by immune tolerance. In conclusion, immune tolerance may reduce proinflammatory cytokine expression and reactive oxygen species production. Our findings provide insights into the effects of endotoxin tolerance on innate immune cells and social behaviors.
Collapse
Affiliation(s)
- Vichuda Charoensaensuk
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Sian-Ting Huang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, 404328, Taiwan
| | - Sheng-Yun Xie
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chao-Wei Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Yen-Chang Chen
- Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Han-Tsung Cheng
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Sheng-Wei Lai
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Kai Shen
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Hui-Jung Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, China Medical University, Taichung, 404328, Taiwan; Laboratory for Neural Repair, China Medical University Hospital, Taichung, 404327, Taiwan; Biomedical Technology R&D Center, China Medical University Hospital, Taichung, 404327, Taiwan.
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
19
|
Li C, Liu R, Xiong Z, Bao X, Liang S, Zeng H, Jin W, Gong Q, Liu L, Guo J. Ferroptosis: a potential target for the treatment of atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:331-344. [PMID: 38327187 PMCID: PMC10984869 DOI: 10.3724/abbs.2024016] [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: 11/06/2023] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Atherosclerosis (AS), the main contributor to acute cardiovascular events, such as myocardial infarction and ischemic stroke, is characterized by necrotic core formation and plaque instability induced by cell death. The mechanisms of cell death in AS have recently been identified and elucidated. Ferroptosis, a novel iron-dependent form of cell death, has been proven to participate in atherosclerotic progression by increasing endothelial reactive oxygen species (ROS) levels and lipid peroxidation. Furthermore, accumulated intracellular iron activates various signaling pathways or risk factors for AS, such as abnormal lipid metabolism, oxidative stress, and inflammation, which can eventually lead to the disordered function of macrophages, vascular smooth muscle cells, and vascular endothelial cells. However, the molecular pathways through which ferroptosis affects AS development and progression are not entirely understood. This review systematically summarizes the interactions between AS and ferroptosis and provides a feasible approach for inhibiting AS progression from the perspective of ferroptosis.
Collapse
Affiliation(s)
- Chengyi Li
- School of MedicineYangtze UniversityJingzhou434020China
| | - Ran Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Zhenyu Xiong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Xue Bao
- School of MedicineYangtze UniversityJingzhou434020China
| | - Sijia Liang
- Department of PharmacologyZhongshan School of MedicineSun Yat-Sen UniversityGuangzhou510120China
| | - Haotian Zeng
- Department of GastroenterologyShenzhen People’s HospitalThe Second Clinical Medical CollegeJinan UniversityShenzhen518000China
| | - Wei Jin
- Department of Second Ward of General PediatricsSuizhou Central HospitalHubei University of MedicineSuizhou441300China
| | - Quan Gong
- School of MedicineYangtze UniversityJingzhou434020China
| | - Lian Liu
- School of MedicineYangtze UniversityJingzhou434020China
| | - Jiawei Guo
- School of MedicineYangtze UniversityJingzhou434020China
| |
Collapse
|
20
|
Shahror RA, Morris CA, Mohammed AA, Wild M, Zaman B, Mitchell CD, Phillips PH, Rusch NJ, Shosha E, Fouda AY. Role of myeloid cells in ischemic retinopathies: recent advances and unanswered questions. J Neuroinflammation 2024; 21:65. [PMID: 38454477 PMCID: PMC10918977 DOI: 10.1186/s12974-024-03058-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Myeloid cells including microglia and macrophages play crucial roles in retinal homeostasis by clearing cellular debris and regulating inflammation. These cells are activated in several blinding ischemic retinal diseases including diabetic retinopathy, where they may exert both beneficial and detrimental effects on neurovascular function and angiogenesis. Myeloid cells impact the progression of retinal pathologies and recent studies suggest that targeting myeloid cells is a promising therapeutic strategy to mitigate diabetic retinopathy and other ischemic retinal diseases. This review summarizes the recent advances in our understanding of the role of microglia and macrophages in retinal diseases and focuses on the effects of myeloid cells on neurovascular injury and angiogenesis in ischemic retinopathies. We highlight gaps in knowledge and advocate for a more detailed understanding of the role of myeloid cells in retinal ischemic injury to fully unlock the potential of targeting myeloid cells as a therapeutic strategy for retinal ischemia.
Collapse
Affiliation(s)
- Rami A Shahror
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Carol A Morris
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Aya A Mohammed
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Melissa Wild
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Bushra Zaman
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Christian D Mitchell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Paul H Phillips
- Department of Ophthalmology, Harvey & Bernice Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Esraa Shosha
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA.
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt.
| |
Collapse
|
21
|
Gargas J, Janowska J, Gebala P, Maksymiuk W, Sypecka J. Reactive Gliosis in Neonatal Disorders: Friend or Foe for Neuroregeneration? Cells 2024; 13:131. [PMID: 38247822 PMCID: PMC10813898 DOI: 10.3390/cells13020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
A developing nervous system is particularly vulnerable to the influence of pathophysiological clues and injuries in the perinatal period. Astrocytes are among the first cells that react to insults against the nervous tissue, the presence of pathogens, misbalance of local tissue homeostasis, and a lack of oxygen and trophic support. Under this background, it remains uncertain if induced astrocyte activation, recognized as astrogliosis, is a friend or foe for progressing neonatal neurodevelopment. Likewise, the state of astrocyte reactivity is considered one of the key factors discriminating between either the initiation of endogenous reparative mechanisms compensating for aberrations in the structures and functions of nervous tissue or the triggering of neurodegeneration. The responses of activated cells are modulated by neighboring neural cells, which exhibit broad immunomodulatory and pro-regenerative properties by secreting a plethora of active compounds (including interleukins and chemokines, neurotrophins, reactive oxygen species, nitric oxide synthase and complement components), which are engaged in cell crosstalk in a paracrine manner. As the developing nervous system is extremely sensitive to the influence of signaling molecules, even subtle changes in the composition or concentration of the cellular secretome can have significant effects on the developing neonatal brain. Thus, modulating the activity of other types of cells and their interactions with overreactive astrocytes might be a promising strategy for controlling neonatal astrogliosis.
Collapse
Affiliation(s)
| | | | | | | | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, A. Pawinskiego 5, 02-106 Warsaw, Poland; (J.G.); (J.J.)
| |
Collapse
|
22
|
Gao X, Su G, Chai M, Shen M, Hu Z, Chen W, Gao J, Li R, Ma T, An Y, Zhang Z. Research progress on mechanisms of ischemic stroke: Regulatory pathways involving Microglia. Neurochem Int 2024; 172:105656. [PMID: 38081419 DOI: 10.1016/j.neuint.2023.105656] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/19/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
Microglia, as the intrinsic immune cells in the brain, are activated following ischemic stroke. Activated microglia participate in the pathological processes after stroke through polarization, autophagy, phagocytosis, pyroptosis, ferroptosis, apoptosis, and necrosis, thereby influencing the injury and repair following stroke. It has been established that polarized M1 and M2 microglia exhibit pro-inflammatory and anti-inflammatory effects, respectively. Autophagy and phagocytosis in microglia following ischemia are dynamic processes, where moderate levels promote cell survival, while excessive responses may exacerbate neurofunctional deficits following stroke. Additionally, pyroptosis and ferroptosis in microglia after ischemic stroke contribute to the release of harmful cytokines, further aggravating the damage to brain tissue due to ischemia. This article discusses the different functional states of microglia in ischemic stroke research, highlighting current research trends and gaps, and provides insights and guidance for further study of ischemic stroke.
Collapse
Affiliation(s)
- Xin Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730030, China
| | - Miao Chai
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Minghui Shen
- Medical Laboratories, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Zhenzhen Hu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Ruixin Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Tianfei Ma
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Yang An
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
23
|
Desmond LW, Holbrook EM, Wright CTO, Zambrano CA, Stamper CE, Bohr AD, Frank MG, Podell BK, Moreno JA, MacDonald AS, Reber SO, Hernández-Pando R, Lowry CA. Effects of Mycobacterium vaccae NCTC 11659 and Lipopolysaccharide Challenge on Polarization of Murine BV-2 Microglial Cells. Int J Mol Sci 2023; 25:474. [PMID: 38203645 PMCID: PMC10779110 DOI: 10.3390/ijms25010474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Previous studies have shown that the in vivo administration of soil-derived bacteria with anti-inflammatory and immunoregulatory properties, such as Mycobacterium vaccae NCTC 11659, can prevent a stress-induced shift toward an inflammatory M1 microglial immunophenotype and microglial priming in the central nervous system (CNS). It remains unclear whether M. vaccae NCTC 11659 can act directly on microglia to mediate these effects. This study was designed to determine the effects of M. vaccae NCTC 11659 on the polarization of naïve BV-2 cells, a murine microglial cell line, and BV-2 cells subsequently challenged with lipopolysaccharide (LPS). Briefly, murine BV-2 cells were exposed to 100 µg/mL whole-cell, heat-killed M. vaccae NCTC 11659 or sterile borate-buffered saline (BBS) vehicle, followed, 24 h later, by exposure to 0.250 µg/mL LPS (Escherichia coli 0111: B4; n = 3) in cell culture media vehicle (CMV) or a CMV control condition. Twenty-four hours after the LPS or CMV challenge, cells were harvested to isolate total RNA. An analysis using the NanoString platform revealed that, by itself, M. vaccae NCTC 11659 had an "adjuvant-like" effect, while exposure to LPS increased the expression of mRNAs encoding proinflammatory cytokines, chemokine ligands, the C3 component of complement, and components of inflammasome signaling such as Nlrp3. Among LPS-challenged cells, M. vaccae NCTC 11659 had limited effects on differential gene expression using a threshold of 1.5-fold change. A subset of genes was assessed using real-time reverse transcription polymerase chain reaction (real-time RT-PCR), including Arg1, Ccl2, Il1b, Il6, Nlrp3, and Tnf. Based on the analysis using real-time RT-PCR, M. vaccae NCTC 11659 by itself again induced "adjuvant-like" effects, increasing the expression of Il1b, Il6, and Tnf while decreasing the expression of Arg1. LPS by itself increased the expression of Ccl2, Il1b, Il6, Nlrp3, and Tnf while decreasing the expression of Arg1. Among LPS-challenged cells, M. vaccae NCTC 11659 enhanced LPS-induced increases in the expression of Nlrp3 and Tnf, consistent with microglial priming. In contrast, among LPS-challenged cells, although M. vaccae NCTC 11659 did not fully prevent the effects of LPS relative to vehicle-treated control conditions, it increased Arg1 mRNA expression, suggesting that M. vaccae NCTC 11659 induces an atypical microglial phenotype. Thus, M. vaccae NCTC 11659 acutely (within 48 h) induced immune-activating and microglial-priming effects when applied directly to murine BV-2 microglial cells, in contrast to its long-term anti-inflammatory and immunoregulatory effects observed on the CNS when whole-cell, heat-killed preparations of M. vaccae NCTC 11659 were given peripherally in vivo.
Collapse
Affiliation(s)
- Luke W. Desmond
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Evan M. Holbrook
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Caelan T. O. Wright
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Cristian A. Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Christopher E. Stamper
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Adam D. Bohr
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
| | - Matthew G. Frank
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Brendan K. Podell
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - Julie A. Moreno
- Prion Research Center, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Center for Healthy Aging, Colorado State University, Fort Collins, CO 80523, USA
| | - Andrew S. MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9NT, UK;
| | - Stefan O. Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional De Ciencias Médicas Y Nutrición Salvador Zubirán, Ciudad de México 14080, Mexico;
| | - Christopher A. Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA; (L.W.D.); (E.M.H.); (C.T.O.W.); (C.A.Z.); (C.E.S.); (A.D.B.); (M.G.F.)
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
- Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
24
|
Ren K, Pei J, Guo Y, Jiao Y, Xing H, Xie Y, Yang Y, Feng Q, Yang J. Regulated necrosis pathways: a potential target for ischemic stroke. BURNS & TRAUMA 2023; 11:tkad016. [PMID: 38026442 PMCID: PMC10656754 DOI: 10.1093/burnst/tkad016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/24/2022] [Indexed: 12/01/2023]
Abstract
Globally, ischemic stroke causes millions of deaths per year. The outcomes of ischemic stroke are largely determined by the amount of ischemia-related and reperfusion-related neuronal death in the infarct region. In the infarct region, cell injuries follow either the regulated pathway involving precise signaling cascades, such as apoptosis and autophagy, or the nonregulated pathway, which is uncontrolled by any molecularly defined effector mechanisms such as necrosis. However, numerous studies have recently found that a certain type of necrosis can be regulated and potentially modified by drugs and is nonapoptotic; this type of necrosis is referred to as regulated necrosis. Depending on the signaling pathway, various elements of regulated necrosis contribute to the development of ischemic stroke, such as necroptosis, pyroptosis, ferroptosis, pathanatos, mitochondrial permeability transition pore-mediated necrosis and oncosis. In this review, we aim to summarize the underlying molecular mechanisms of regulated necrosis in ischemic stroke and explore the crosstalk and interplay among the diverse types of regulated necrosis. We believe that targeting these regulated necrosis pathways both pharmacologically and genetically in ischemia-induced neuronal death and protection could be an efficient strategy to increase neuronal survival and regeneration in ischemic stroke.
Collapse
Affiliation(s)
- Kaidi Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Yuanyuan Guo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yuxue Jiao
- Quality Management Department, Henan No. 3 Provincial People’s Hospital, Henan No. 3 Provincial People’s Hospital, Zhengzhou 450052, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yi Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Research Center for Clinical System Biology, Translational Medicine Center, No. 1 Jianshe Dong Road, ErQi District, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Feng
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Province Research Center for Kidney Disease, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
| | - Jing Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou 450052, China
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, No. 1 Jianshe Dong Road, ErQi District, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
25
|
Li L, He G, Shi M, Zhu J, Cheng Y, Chen Y, Chen J, Xue Q. Edaravone dexborneol ameliorates cognitive impairment by regulating the NF-κB pathway through AHR and promoting microglial polarization towards the M2 phenotype in mice with bilateral carotid artery stenosis (BCAS). Eur J Pharmacol 2023; 957:176036. [PMID: 37673366 DOI: 10.1016/j.ejphar.2023.176036] [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: 03/31/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Cerebral small vessel disease (CSVD) is one of the most important causes of stroke and vascular dementia, so exploring effective treatment modalities for CSVD is warranted. This study aimed to explore the anti-inflammatory effects of Edaravone dexborneol (C.EDA) in a CSVD model. Mice with CSVD showed distinct cognitive decline, as assessed by the Morris water maze (MWM). Pathological staining verified leakage across the blood‒brain barrier (BBB), microglial proliferation, neuronal loss and demyelination. Western blot analysis demonstrated that M1 microglia dominated prophase and released proinflammatory molecules; the aryl hydrocarbon receptor (AHR) was found to participate in modulating nuclear factor-kappa B (NF-κB) signalling activation through tumour necrosis factor receptor-associated factor-6 (TRAF6). C.EDA treatment resulted in the polarization of microglia from the M1 to the M2 phenotype. Mice sequentially treated with C.EDA exhibited a significant improvement in cognitive function; expression of the anti-inflammatory cytokines and modulatory proteins AHR and TRAF6 was upregulated, while the levels of pNF-κBp65 and pIΚBα were downregulated. C.EDA promoted microglial activation towards the M2 phenotype by upregulating AHR expression, which prevented TRAF6 ubiquitination, promoted NF-κB RelA/p65 protein degradation and inhibited subsequent NF-κB phosphorylation. Mechanistically, the anti-inflammatory effect of C.EDA alleviated neuronal loss and myelin damage, while at the functional level, C.EDA improved cognitive function and thus showed good application prospects.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China; Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Guojun He
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Mingyu Shi
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China
| | - Yongqing Cheng
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Yang Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Jin Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
26
|
Fang S, Wu Z, Guo Y, Zhu W, Wan C, Yuan N, Chen J, Hao W, Mo X, Guo X, Fan L, Li X, Chen J. Roles of microglia in adult hippocampal neurogenesis in depression and their therapeutics. Front Immunol 2023; 14:1193053. [PMID: 37881439 PMCID: PMC10597707 DOI: 10.3389/fimmu.2023.1193053] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Adult hippocampal neurogenesis generates functional neurons from neural progenitor cells in the hippocampal dentate gyrus (DG) to complement and repair neurons and neural circuits, thus benefiting the treatment of depression. Increasing evidence has shown that aberrant microglial activity can disrupt the appropriate formation and development of functional properties of neurogenesis, which will play a crucial role in the occurrence and development of depression. However, the mechanisms of the crosstalk between microglia and adult hippocampal neurogenesis in depression are not yet fully understood. Therefore, in this review, we first introduce recent discoveries regarding the roles of microglia and adult hippocampal neurogenesis in the etiology of depression. Then, we systematically discuss the possible mechanisms of how microglia regulate adult hippocampal neurogenesis in depression according to recent studies, which involve toll-like receptors, microglial polarization, fractalkine-C-X3-C motif chemokine receptor 1, hypothalamic-pituitary-adrenal axis, cytokines, brain-derived neurotrophic factor, and the microbiota-gut-brain axis, etc. In addition, we summarize the promising drugs that could improve the adult hippocampal neurogenesis by regulating the microglia. These findings will help us understand the complicated pathological mechanisms of depression and shed light on the development of new treatment strategies for this disease.
Collapse
Affiliation(s)
- Shaoyi Fang
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhibin Wu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yali Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wenjun Zhu
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Chunmiao Wan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Shenzhen People’s Hospital, 2Clinical Medical College, Jinan University, Shenzhen, China
| | - Jianbei Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenzhi Hao
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaowei Mo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaofang Guo
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lili Fan
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
Zong B, Yu F, Zhang X, Pang Y, Zhao W, Sun P, Li L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res Rev 2023; 90:102026. [PMID: 37532007 DOI: 10.1016/j.arr.2023.102026] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Since the discovery of the mechanosensitive Piezo1 channel in 2010, there has been a significant amount of research conducted to explore its regulatory role in the physiology and pathology of various organ systems. Recently, a growing body of compelling evidence has emerged linking the activity of the mechanosensitive Piezo1 channel to health and disease of the central nervous system. However, the exact mechanisms underlying these associations remain inadequately comprehended. This review systematically summarizes the current research on the mechanosensitive Piezo1 channel and its implications for central nervous system mechanobiology, retrospects the results demonstrating the regulatory role of the mechanosensitive Piezo1 channel on various cell types within the central nervous system, including neural stem cells, neurons, oligodendrocytes, microglia, astrocytes, and brain endothelial cells. Furthermore, the review discusses the current understanding of the involvement of the Piezo1 channel in central nervous system disorders, such as Alzheimer's disease, multiple sclerosis, glaucoma, stroke, and glioma.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Fengzhi Yu
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyou Zhang
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Yige Pang
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, Zhejiang, China
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
28
|
Sheu ML, Pan LY, Yang CN, Sheehan J, Pan LY, You WC, Wang CC, Chen HS, Pan HC. Neuronal Death Caused by HMGB1-Evoked via Inflammasomes from Thrombin-Activated Microglia Cells. Int J Mol Sci 2023; 24:12664. [PMID: 37628850 PMCID: PMC10454604 DOI: 10.3390/ijms241612664] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Microglial cells are a macrophage-like cell type residing within the CNS. These cells evoke pro-inflammatory responses following thrombin-induced brain damage. Inflammasomes, which are large caspase-1-activating protein complexes, play a critical role in mediating the extracellular release of HMGB1 in activated immune cells. The exact role of inflammasomes in microglia activated by thrombin remains unclear, particularly as it relates to the downstream functions of HMGB1. After receiving microinjections of thrombin, Sprague Dawley rats of 200 to 250 gm were studied in terms of behaviors and immunohistochemical staining. Primary culture of microglia cells and BV-2 cells were used for the assessment of signal pathways. In a water maze test and novel object recognition analysis, microinjections of thrombin impaired rats' short-term and long-term memory, and such detrimental effects were alleviated by injecting anti-HMGB-1 antibodies. After thrombin microinjections, the increased oxidative stress of neurons was aggravated by HMGB1 injections but attenuated by anti-HMGB-1 antibodies. Such responses occurred in parallel with the volume of activated microglia cells, as well as their expressions of HMGB-1, IL-1β, IL-18, and caspase-I. In primary microglia cells and BV-2 cell lines, thrombin also induced NO release and mRNA expressions of iNOS, IL-1β, IL-18, and activated caspase-I. HMGB-1 aggravated these responses, which were abolished by anti-HMGB-1 antibodies. In conclusion, thrombin induced microglia activation through triggering inflammasomes to release HMGB1, contributing to neuronal death. Such an action was counteracted by the anti-HMGB-1 antibodies. The refinement of HMGB-1 modulated the neuro-inflammatory response, which was attenuated in thrombin-associated neurodegenerative disorder.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei 106319, Taiwan;
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA;
| | - Liang-Yu Pan
- Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Weir-Chiang You
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 40210, Taiwan;
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan;
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs’ Taichung Metro-Harbor Hospital, Taichung 40210, Taiwan;
| | - Hung-Chuan Pan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| |
Collapse
|
29
|
Liu L, Han L, Dong L, He Z, Gao K, Chen X, Guo JC, Zhao Y. The hypoxia-associated genes in immune infiltration and treatment options of lung adenocarcinoma. PeerJ 2023; 11:e15621. [PMID: 37576511 PMCID: PMC10414028 DOI: 10.7717/peerj.15621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/01/2023] [Indexed: 08/15/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a common lung cancer with a poor prognosis under standard chemotherapy. Hypoxia is a crucial factor in the development of solid tumors, and hypoxia-related genes (HRGs) are closely associated with the proliferation of LUAD cells. Methods In this study, LUAD HRGs were screened, and bioinformatics analysis and experimental validation were conducted. The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases were used to gather LUAD RNA-seq data and accompanying clinical information. LUAD subtypes were identified by unsupervised cluster analysis, and immune infiltration analysis of subtypes was conducted by GSVA and ssGSEA. Cox regression and LASSO regression analyses were used to obtain prognosis-related HRGs. Prognostic analysis was used to evaluate HRGs. Differences in enrichment pathways and immunotherapy were observed between risk groups based on GSEA and the TIDE method. Finally, RT-PCR and in vitro experiments were used to confirm prognosis-related HRG expression in LUAD cells. Results Two hypoxia-associated subtypes of LUAD were distinguished, demonstrating significant differences in prognostic analysis and immunological characteristics between subtypes. A prognostic model based on six HRGs (HK1, PDK3, PFKL, SLC2A1, STC1, and XPNPEP1) was developed for LUAD. HK1, SLC2A1, STC1, and XPNPEP1 were found to be risk factors for LUAD. PDK3 and PFKL were protective factors in LUAD patients. Conclusion This study demonstrates the effect of hypoxia-associated genes on immune infiltration in LUAD and provides options for immunotherapy and therapeutic strategies in LUAD.
Collapse
Affiliation(s)
- Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lina Han
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zihao He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Cheng Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- The Research Center for Ubiquitous Computing Systems (CUbiCS), Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Chen Y, Peng F, Yang C, Hou H, Xing Z, Chen J, Liu L, Peng C, Li D. SIRT1 activation by 2,3,5,6-tetramethylpyrazine alleviates neuroinflammation via inhibiting M1 microglia polarization. Front Immunol 2023; 14:1206513. [PMID: 37600790 PMCID: PMC10436537 DOI: 10.3389/fimmu.2023.1206513] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Background Neuroinflammation has been reported as a potential contributing factor to brain diseases, and is characterized by activated microglia with release of multiple inflammatory mediators. 2,3,5,6-Tetramethylpyrazine (TMP) is an active alkaloid in Ligusticum chuanxiong Hort. and has various biological activities, including anti-inflammatory and neuroprotection properties. However, the anti-neuroinflammatory activity of TMP has been less studied and its potential molecular mechanisms in this field remain unclear. This study aimed to investigate the effects of TMP and its underlying mechanisms in neuroinflammation. Methods In vitro, lipopolysaccharide (LPS)-stimulated BV2 microglia were used to assess the effects of TMP on inflammatory cytokines as well as the components of the SIRT1/NF-κB signaling pathway, which were measured by using ELISA, western blotting, qRT-qPCR and immunofluorescence. Moreover, LPS-induced acute neuroinflammation model in mice was performed to detect whether TMP could exert anti-neuroinflammatory effects in vivo, and the EX527, a SIRT1 inhibitor, were given intraperitoneally every two days prior to TMP treatment. Serums and spinal trigeminal nucleus (Sp5) tissues were collected for ELISA assay, and the Sp5 tissues were used for HE staining, Nissl staining, immunofluorescence, qRT-PCR and western blotting. Results In vitro, TMP treatment significantly reduced the secretion of pro-inflammatory cytokines, including TNF-α and IL-6, promoted SIRT1 protein expression and inactivated NF-κB signaling pathway in LPS-induced neuroinflammation. Interestingly, pretreatment with EX527 blocked the therapeutic effects of TMP on neuroinflammation in vitro. Furthermore, TMP reduced the levels of pro-inflammatory cytokines and chemokines, and prevented microglia from polarizing towards a pro-inflammatory state through activating SIRT1 and inhibiting NF-κB activation in LPS-induced neuroinflammation in mice. And EX527 reversed the beneficial effects of TMP against LPS exposure in mice. Conclusion In summary, this study unravels that TMP could mitigate LPS-induced neuroinflammation via SIRT1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation and Application, Zhejiang Ocean University, Zhoushan, Zhejiang, China
| | - Huan Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junren Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Liu
- Chiatai Qingchunbao Pharmaceutical Co., Ltd., Hangzhou, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
31
|
Huang H, Li S, Zhang Y, He C, Hua Z. Microglial Priming in Bilirubin-Induced Neurotoxicity. Neurotox Res 2023; 41:338-348. [PMID: 37058197 DOI: 10.1007/s12640-023-00643-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/05/2023] [Accepted: 04/02/2023] [Indexed: 04/15/2023]
Abstract
Neuroinflammation is a major contributor to bilirubin-induced neurotoxicity, which results in severe neurological deficits. Microglia are the primary immune cells in the brain, with M1 microglia promoting inflammatory injury and M2 microglia inhibiting neuroinflammation. Controlling microglial inflammation could be a promising therapeutic strategy for reducing bilirubin-induced neurotoxicity. Primary microglial cultures were prepared from 1-3-day-old rats. In the early stages of bilirubin treatment, pro-/anti-inflammatory (M1/M2) microglia mixed polarization was observed. In the late stages, bilirubin persistence induced dominant proinflammatory microglia, forming an inflammatory microenvironment and inducing iNOS expression as well as the release of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β. Simultaneously, nuclear factor-kappa B (NF-κB) was activated and translocated into the nucleus, upregulating inflammatory target genes. As well known, neuroinflammation can have an effect on N-methyl-D-aspartate receptor (NMDAR) expression or function, which is linked to cognition. Treatment with bilirubin-treated microglia-conditioned medium did affect the expression of IL-1β, NMDA receptor subunit 2A (NR2A), and NMDA receptor subunit 2B (NR2B) in neurons. However, VX-765 effectively reduces the levels of proinflammatory cytokines TNF-α, IL-6, and IL-1β, as well as the expressions of CD86, and increases the expressions of anti-inflammatory related Arg-1. A timely reduction in proinflammatory microglia could protect against bilirubin-induced neurotoxicity.
Collapse
Affiliation(s)
- Hongmei Huang
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Siyu Li
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Yan Zhang
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Chunmei He
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Ziyu Hua
- Department of Neonatology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
- Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China.
| |
Collapse
|
32
|
Sheu ML, Pan LY, Yang CN, Sheehan J, Pan LY, You WC, Wang CC, Pan HC. Thrombin-Induced Microglia Activation Modulated through Aryl Hydrocarbon Receptors. Int J Mol Sci 2023; 24:11416. [PMID: 37511175 PMCID: PMC10380349 DOI: 10.3390/ijms241411416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Thrombin is a multifunctional serine protein which is closely related to neurodegenerative disorders. The Aryl hydrocarbon receptor (AhR) is well expressed in microglia cells involving inflammatory disorders of the brain. However, it remains unclear as to how modulation of AhR expression by thrombin is related to the development of neurodegeneration disorders. In this study, we investigated the role of AhR in the development of thrombin-induced neurodegenerative processes, especially those concerning microglia. The primary culture of either wild type or AhR deleted microglia, as well as BV-2 cell lines, was used for an in vitro study. Hippocampal slice culture and animals with either wild type or with AhR deleted were used for the ex vivo and in vivo studies. Simulations of ligand protein docking showed a strong integration between the thrombin and AhR. In thrombin-triggered microglia cells, deleting AhR escalated both the NO release and iNOS expression. Such effects were abolished by the administration of the AhR agonist. In thrombin-activated microglia cells, downregulating AhR increased the following: vascular permeability, pro-inflammatory genetic expression, MMP-9 activity, and the ratio of M1/M2 phenotype. In the in vivo study, thrombin induced the activation of microglia and their volume, thereby contributing to the deterioration of neurobehavior. Deleting AhR furthermore aggravated the response in terms of impaired neurobehavior, increasing brain edema, aggregating microglia, and increasing neuronal death. In conclusion, thrombin caused the activation of microglia through increased vessel permeability, expression of inflammatory response, and phenotype of M1 microglia, as well the MMP activity. Deleting AhR augmented the above detrimental effects. These findings indicate that the modulation of AhR is essential for the regulation of thrombin-induced brain damages and that the AhR agonist may harbor the potentially therapeutic effect in thrombin-induced neurodegenerative disorder.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei 106319, Taiwan;
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA;
| | - Liang-Yu Pan
- Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Weir-Chiang You
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 40210, Taiwan;
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan;
| | - Hung-Chuan Pan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| |
Collapse
|
33
|
Moss CE, Phipps H, Wilson HL, Kiss-Toth E. Markers of the ageing macrophage: a systematic review and meta-analysis. Front Immunol 2023; 14:1222308. [PMID: 37520567 PMCID: PMC10373068 DOI: 10.3389/fimmu.2023.1222308] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Ageing research is establishing macrophages as key immune system regulators that undergo functional decline. Due to heterogeneity between species and tissue populations, a plethora of data exist and the power of scientific conclusions can vary substantially. This meta-analysis by information content (MAIC) and systematic literature review (SLR) aims to determine overall changes in macrophage gene and protein expression, as well as function, with age. Methods PubMed was utilized to collate peer-reviewed literature relating to macrophage ageing. Primary studies comparing macrophages in at least two age groups were included. Data pertaining to gene or protein expression alongside method used were extracted for MAIC analysis. For SLR analysis, data included all macrophage-specific changes with age, as well as species, ontogeny and age of groups assessed. Results A total of 240 studies were included; 122 of which qualified for MAIC. The majority of papers focussed on changes in macrophage count/infiltration as a function of age, followed by gene and protein expression. The MAIC found iNOS and TNF to be the most commonly investigated entities, with 328 genes and 175 proteins showing consistent dysregulation with age across the literature. Overall findings indicate that cytokine secretion and phagocytosis are reduced and reactive oxygen species production is increased in the ageing macrophage. Discussion Collectively, our analysis identifies critical regulators in macrophage ageing that are consistently dysregulated, highlighting a plethora of targets for further investigation. Consistent functional changes with age found here can be used to confirm an ageing macrophage phenotype in specific studies and experimental models.
Collapse
Affiliation(s)
- Charlotte E. Moss
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| | - Hew Phipps
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Heather L. Wilson
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- Healthy Lifespan Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
34
|
Huang ZP, Liu SF, Zhuang JL, Li LY, Li MM, Huang YL, Chen YH, Chen XR, Lin S, Ye LC, Chen CN. Role of microglial metabolic reprogramming in Parkinson's disease. Biochem Pharmacol 2023; 213:115619. [PMID: 37211170 DOI: 10.1016/j.bcp.2023.115619] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder characterized by damage to nigrostriatal dopaminergic neurons. Key pathogenic mechanisms underlying PD include alpha-synuclein misfolding and aggregation, impaired protein clearance, mitochondrial dysfunction, oxidative stress, and neuroinflammation. However, to date, no study has confirmed the specific pathogenesis of PD. Similarly, current PD treatment methods still have shortcomings. Although some emerging therapies have proved effective for PD, the specific mechanism still needs further clarification. Metabolic reprogramming, a term first proposed by Warburg, is applied to the metabolic energy characteristics of tumor cells. Microglia have similar metabolic characteristics. Pro-inflammatory M1 type and anti-inflammatory M2 type are the two types of activated microglia, which exhibit different metabolic patterns in glucose, lipid, amino acid, and iron metabolism. Additionally, mitochondrial dysfunction may be involved in microglial metabolic reprogramming by activating various signaling mechanisms. Functional changes in microglia resulting from metabolic reprogramming can cause changes in the brain microenvironment, thus playing an important role in neuroinflammation or tissue repair. The involvement of microglial metabolic reprogramming in PD pathogenesis has been confirmed. Neuroinflammation and dopaminergic neuronal death can effectively be reduced by inhibiting certain metabolic pathways in M1 microglia or reverting M1 cells to the M2 phenotype. This review summarizes the relationship between microglial metabolic reprogramming and PD and provides strategies for PD treatment.
Collapse
Affiliation(s)
- Zheng-Ping Huang
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Shu-Fen Liu
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Jian-Long Zhuang
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou, China
| | - Lin-Yi Li
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Mi-Mi Li
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Ya-Li Huang
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China
| | - Yan-Hong Chen
- Department of Neurology, Shishi General Hospital, Quanzhou, Fujian Province 362000, China
| | - Xiang-Rong Chen
- Department of Neurosurgery, Second Affiliated Hospital, Second Clinical Medical College, Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Center of Neurological and Metabolic Research, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province 362000, China; Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Li-Chao Ye
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China.
| | - Chun-Nuan Chen
- Department of Neurology, Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province 362000, China.
| |
Collapse
|
35
|
Wang Y, Qin WY, Wang Q, Liu XN, Li XH, Ye XQ, Bai Y, Zhang Y, Liu P, Wang XL, Zhou YH, Shao ZB, Yuan HP. Young Sca-1 + bone marrow stem cell-derived exosomes preserve visual function via the miR-150-5p/MEKK3/JNK/c-Jun pathway to reduce M1 microglial polarization. J Nanobiotechnology 2023; 21:194. [PMID: 37322478 DOI: 10.1186/s12951-023-01944-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Polarization of microglia, the resident retinal immune cells, plays important roles in mediating both injury and repair responses post-retinal ischemia-reperfusion (I/R) injury, which is one of the main pathological mechanisms behind ganglion cell apoptosis. Aging could perturb microglial balances, resulting in lowered post-I/R retinal repair. Young bone marrow (BM) stem cell antigen 1-positive (Sca-1+) cells have been demonstrated to have higher reparative capabilities post-I/R retinal injury when transplanted into old mice, where they were able to home and differentiate into retinal microglia. METHODS Exosomes were enriched from young Sca-1+ or Sca-1- cells, and injected into the vitreous humor of old mice post-retinal I/R. Bioinformatics analyses, including miRNA sequencing, was used to analyze exosome contents, which was confirmed by RT-qPCR. Western blot was then performed to examine expression levels of inflammatory factors and underlying signaling pathway proteins, while immunofluorescence staining was used to examine the extent of pro-inflammatory M1 microglial polarization. Fluoro-Gold labelling was then utilized to identify viable ganglion cells, while H&E staining was used to examine retinal morphology post-I/R and exosome treatment. RESULTS Sca-1+ exosome-injected mice yielded better visual functional preservation and lowered inflammatory factors, compared to Sca-1-, at days 1, 3, and 7 days post-I/R. miRNA sequencing found that Sca-1+ exosomes had higher miR-150-5p levels, compared to Sca-1- exosomes, which was confirmed by RT-qPCR. Mechanistic analysis found that miR-150-5p from Sca-1+ exosomes repressed the mitogen-activated protein kinase kinase kinase 3 (MEKK3)/JNK/c-Jun axis, leading to IL-6 and TNF-α downregulation, and subsequently reduced microglial polarization, all of which contributes to reduced ganglion cell apoptosis and preservation of proper retinal morphology. CONCLUSION This study elucidates a potential new therapeutic approach for neuroprotection against I/R injury, via delivering miR-150-5p-enriched Sca-1+ exosomes, which targets the miR-150-5p/MEKK3/JNK/c-Jun axis, thereby serving as a cell-free remedy for treating retinal I/R injury and preserving visual functioning.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan-Yun Qin
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Harbin, China
| | - Xin-Na Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Harbin, China
| | - Xiang-Hui Li
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin-Qi Ye
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Bai
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pan Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin-Lin Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Hang Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zheng-Bo Shao
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Hui-Ping Yuan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
36
|
Xiong S, Su X, Kang Y, Si J, Wang L, Li X, Ma K. Effect and mechanism of chlorogenic acid on cognitive dysfunction in mice by lipopolysaccharide-induced neuroinflammation. Front Immunol 2023; 14:1178188. [PMID: 37292216 PMCID: PMC10244504 DOI: 10.3389/fimmu.2023.1178188] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/11/2023] [Indexed: 06/10/2023] Open
Abstract
Background Neuroinflammation is an important factor causing numerous neurodegenerative pathologies. Inflammation can lead to abnormal neuronal structure and function and even death, followed by cognitive dysfunction. There is growing evidence that chlorogenic acid has anti-inflammatory effects and immunomodulatory activity. Purpose The aim of this study was to elucidate the potential targets and molecular mechanisms of chlorogenic acid in the treatment of neuroinflammation. Methods We used the lipopolysaccharide-induced neuroinflammation mouse model and the lipopolysaccharide-stimulated BV-2 cells in vitro model. Behavioral scores and experiments were used to assess cognitive dysfunction in mice. HE staining and immunohistochemistry were used to assess neuronal damage in the mouse brain. Immunofluorescence detected microglia polarization in mouse brain. Western blot and flow cytometry detected the polarization of BV-2 cells. The migration of BV-2 cells was detected by wound healing assay and transwell assay. Potential targets for chlorogenic acid to exert protective effects were predicted by network pharmacology. These targets were then validated using molecular docking and experiments. Results The results of in vivo experiments showed that chlorogenic acid had an obvious ameliorating effect on neuroinflammation-induced cognitive dysfunction. We found that chlorogenic acid was able to inhibit BV-2 cells M1 polarization and promote BV-2 cells M2 polarization in vitro while also inhibiting the abnormal migration of BV-2 cells. Based on the network pharmacology results, we identified the TNF signaling pathway as a key signaling pathway in which chlorogenic acid exerts anti-neuroinflammatory effects. Among them, Akt1, TNF, MMP9, PTGS2, MAPK1, MAPK14, and RELA are the core targets for chlorogenic acid to function. Conclusion Chlorogenic acid can inhibit microglial polarization toward the M1 phenotype and improve neuroinflammation-induced cognitive dysfunction in mice by modulating these key targets in the TNF signaling pathway.
Collapse
Affiliation(s)
- Siyuan Xiong
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Xuyang Su
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Yingjie Kang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Junqiang Si
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| | - Lu Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pharmacology and Clinical Pharmacy, Shihezi University School of Pharmacy, Shihezi, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, China
- National Health Commission (NHC) Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi, China
| |
Collapse
|
37
|
Kim B, Kim R, Kim HJ, Kim Y, Park SJ, Lee EH, Kim J, Kim J, Choi JW, Park JH, Park KD. Optimization and evaluation of pyridinyl vinyl sulfones as Nrf2 activator for the antioxidant and anti-inflammatory effects. Eur J Med Chem 2023; 256:115433. [PMID: 37187090 DOI: 10.1016/j.ejmech.2023.115433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023]
Abstract
Many studies have reported that chalcone-based compounds exhibit biological activities such as anticancer, antioxidant, anti-inflammatory and neuroprotective effects. Among the published chalcone derivatives, (E)-1-(3-methoxypyridin-2-yl)-3-(2-(trifluoromethyl)phenyl)prop-2-en-1-one (VEDA-1209), which is currently undergoing preclinical study, was selected as a starting compound for the development of new nuclear factor erythroid 2-related factor 2 (Nrf2) activators. Based on our previous knowledge, we attempted to redesign and synthesize VEDA-1209 derivatives by introducing the pyridine ring and sulfone moiety to ameliorate its Nrf2 efficacy and drug-like properties. Among the synthesized compounds, (E)-3-chloro-2-(2-((3-methoxypyridin-2-yl)sulfonyl)vinyl) pyridine (10e) was found to have approximately 16-folds higher Nrf2 activating effects than VEDA-1209 (10e: EC50 = 37.9 nM vs VEDA-1209: EC50 = 625 nM) in functional cell-based assay. In addition, 10e effectively improved drug-like properties such as CYP inhibition probability and metabolic stability. Finally, 10e demonstrated excellent antioxidant and anti-inflammatory effects in BV-2 microglial cells and significantly restored spatial memory deficits in lipopolysaccharide (LPS)-induced neuroinflammatory mouse models.
Collapse
Affiliation(s)
- Byungeun Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Rium Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Hyeon Jeong Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoowon Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sun Jun Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Cureverse Co., Ltd., KIST, 1st Floor, H2 Building, Seoul, 02792, Republic of Korea
| | - Elijah Hwejin Lee
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Jushin Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaehwan Kim
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Ji Won Choi
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Cureverse Co., Ltd., KIST, 1st Floor, H2 Building, Seoul, 02792, Republic of Korea.
| | - Jong-Hyun Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Ki Duk Park
- Center for Brain Disorders, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
38
|
Dos Santos BL, Dos Santos CC, Soares JRP, da Silva KC, de Oliveira JVR, Pereira GS, de Araújo FM, Costa MDFD, David JM, da Silva VDA, Butt AM, Costa SL. The Flavonoid Agathisflavone Directs Brain Microglia/Macrophages to a Neuroprotective Anti-Inflammatory and Antioxidant State via Regulation of NLRP3 Inflammasome. Pharmaceutics 2023; 15:pharmaceutics15051410. [PMID: 37242652 DOI: 10.3390/pharmaceutics15051410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Agathisflavone, purified from Cenostigma pyramidale (Tul.) has been shown to be neuroprotective in in vitro models of glutamate-induced excitotoxicity and inflammatory damage. However, the potential role of microglial regulation by agathisflavone in these neuroprotective effects is unclear. Here we investigated the effects of agathisflavone in microglia submitted to inflammatory stimulus in view of elucidating mechanisms of neuroprotection. Microglia isolated from cortices of newborn Wistar rats were exposed to Escherichia coli lipopolysaccharide (LPS, 1 µg/mL) and treated or not with agathisflavone (1 µM). Neuronal PC12 cells were exposed to a conditioned medium from microglia (MCM) treated or not with agathisflavone. We observed that LPS induced microglia to assume an activated inflammatory state (increased CD68, more rounded/amoeboid phenotype). However, most microglia exposed to LPS and agathisflavone, presented an anti-inflammatory profile (increased CD206 and branched-phenotype), associated with the reduction in NO, GSH mRNA for NRLP3 inflammasome, IL1-β, IL-6, IL-18, TNF, CCL5, and CCL2. Molecular docking also showed that agathisflavone bound at the NLRP3 NACTH inhibitory domain. Moreover, in PC12 cell cultures exposed to the MCM previously treated with the flavonoid most cells preserved neurites and increased expression of β-tubulin III. Thus, these data reinforce the anti-inflammatory activity and the neuroprotective effect of agathisflavone, effects associated with the control of NLRP3 inflammasome, standing out it as a promising molecule for the treatment or prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Balbino Lino Dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
- College of Nursing, Federal University of Vale do São Francisco, Petrolina 56304-917, Pernambuco, Brazil
| | - Cleonice Creusa Dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Janaina R P Soares
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Karina C da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Juciele Valeria R de Oliveira
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Gabriele S Pereira
- Group of Studies and Research for Health Development, University Salvador, Salvador 40140-110, Bahia, Brazil
| | - Fillipe M de Araújo
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
- Group of Studies and Research for Health Development, University Salvador, Salvador 40140-110, Bahia, Brazil
| | - Maria de Fátima D Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Jorge Mauricio David
- Department of General and Inorganic Chemistry, Institute of Chemistry, University Federal da Bahia, Salvador 40170-110, Bahia, Brazil
| | - Victor Diogenes A da Silva
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| | - Arthur Morgan Butt
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Av. Reitor Miguel Calmon S/N, Salvador 40231-300, Bahia, Brazil
| |
Collapse
|
39
|
Baicalein ameliorates Alzheimer's disease via orchestration of CX3CR1/NF-κB pathway in a triple transgenic mouse model. Int Immunopharmacol 2023; 118:109994. [PMID: 37098656 DOI: 10.1016/j.intimp.2023.109994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/15/2023]
Abstract
Alzheimer's disease (AD) is a common chronic neurodegenerative disease. Some studies have suggested that dysregulation of microglia activation and the resulting neuroinflammation play an important role in the development of AD pathology. Activated microglia have both M1 and M2 phenotypes and inhibition of M1 phenotype while stimulating M2 phenotype has been considered as a potential treatment for neuroinflammation-related diseases. Baicalein is a class of flavonoids with anti-inflammatory, antioxidant and other biological activities, but its role in AD and the regulation of microglia are limited. The purpose of this study was to investigate the effect of baicalein on the activation of microglia in AD model mice and the related molecular mechanism. Our results showed that baicalein significantly improved the learning and memory ability and AD-related pathology of 3 × Tg-AD mice, inhibited the level of pro-inflammatory factors TNF-α, IL-1β and IL-6, promoted the production of anti-inflammatory factors IL-4 and IL-10, and regulated the microglia phenotype through CX3CR1/NF-κB signaling pathway. In conclusion, baicalein can regulate the phenotypic transformation of activated microglia and reduce neuroinflammation through CX3CR1/NF-κB pathway, thereby improving the learning and memory ability of 3 × Tg-AD mice.
Collapse
|
40
|
Nakagawa Y, Yamada S. Alterations in Brain Neural Network and Stress System in Atopic Dermatitis: Novel Therapeutic Interventions. J Pharmacol Exp Ther 2023; 385:78-87. [PMID: 36828629 DOI: 10.1124/jpet.122.001482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/02/2023] [Accepted: 02/03/2023] [Indexed: 02/26/2023] Open
Abstract
Atopic dermatitis is a common chronic inflammatory skin disease, with most cases experiencing skin barrier dysfunction and enhanced allergen entry, accompanied by cytokine production which evokes predominantly type-2-skewed immune responses, itch, and scratching behavior. Although intense itch and excessive scratching behavior affect progression of skin lesions, it is unclear what causes them. Data suggest that scratching behavior stimulates brain dopaminergic reward and habit learning systems, strengthening habitual scratching behavior, while nocturnal scratching behavior presumably increases locus coeruleus-noradrenergic system activity, prompting sleep disturbances. At the early stage of atopic dermatitis, increased cortisol levels, due to hypothalamic-pituitary-adrenal axis overactivation caused by such system stimulation, can induce dorsolateral prefrontal cortex disturbance with reinforcement of habitual scratching behavior and may aggravate type-2-skewed immune responses in the skin. During the later phases, whereas blunted hypothalamic-pituitary-adrenal axis function and the shift of type-2-dominated to type-1-co-dominated inflammation are induced, noradrenergic system overactivation-associated dorsolateral prefrontal cortex disruption is ongoing and responsible for itch cognitive distortion to catastrophize about itch, which leads to a vicious spiral along with habitual scratching behavior and skin lesions. Data are presented in this review indicating that while skin immune system dysfunction initiates pathologic changes in atopic dermatitis, brain neural network and stress system alterations can promote the progression of this condition. It is also suggested that cognitive distortion contributes to pathology in atopic dermatitis as with some psychiatric disorders and chronic pain. The proposed mechanistic model could lead to development of novel medications for slowing or terminating the relentless progression of this disorder. SIGNIFICANCE STATEMENT: Although conventional pharmacological interventions focusing on skin homeostasis and itch occurrence significantly attenuate clinical signs in atopic dermatitis patients, achievement of 100% improvement is less than 40% in several double-blind, randomized, placebo-controlled trials. Our model predicts that itch cognitive distortion, due to dorsolateral prefrontal cortex disturbance, can significantly contribute to the progression of atopic dermatitis and that agents capable of improving brain neural network, stress system, and skin homeostasis may be effective as interventions in the treatment of this condition.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| |
Collapse
|
41
|
Nakagawa Y, Yamada S. The Relationships Among Metal Homeostasis, Mitochondria, and Locus Coeruleus in Psychiatric and Neurodegenerative Disorders: Potential Pathogenetic Mechanism and Therapeutic Implications. Cell Mol Neurobiol 2023; 43:963-989. [PMID: 35635600 PMCID: PMC11414457 DOI: 10.1007/s10571-022-01234-3] [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: 01/09/2022] [Accepted: 05/15/2022] [Indexed: 11/03/2022]
Abstract
While alterations in the locus coeruleus-noradrenergic system are present during early stages of neuropsychiatric disorders, it is unclear what causes these changes and how they contribute to other pathologies in these conditions. Data suggest that the onset of major depressive disorder and schizophrenia is associated with metal dyshomeostasis that causes glial cell mitochondrial dysfunction and hyperactivation in the locus coeruleus. The effect of the overactive locus coeruleus on the hippocampus, amygdala, thalamus, and prefrontal cortex can be responsible for some of the psychiatric symptoms. Although locus coeruleus overactivation may diminish over time, neuroinflammation-induced alterations are presumably ongoing due to continued metal dyshomeostasis and mitochondrial dysfunction. In early Alzheimer's and Parkinson's diseases, metal dyshomeostasis and mitochondrial dysfunction likely induce locus coeruleus hyperactivation, pathological tau or α-synuclein formation, and neurodegeneration, while reduction of glymphatic and cerebrospinal fluid flow might be responsible for β-amyloid aggregation in the olfactory regions before the onset of dementia. It is possible that the overactive noradrenergic system stimulates the apoptosis signaling pathway and pathogenic protein formation, leading to further pathological changes which can occur in the presence or absence of locus coeruleus hypoactivation. Data are presented in this review indicating that although locus coeruleus hyperactivation is involved in pathological changes at prodromal and early stages of these neuropsychiatric disorders, metal dyshomeostasis and mitochondrial dysfunction are critical factors in maintaining ongoing neuropathology throughout the course of these conditions. The proposed mechanistic model includes multiple pharmacological sites that may be targeted for the treatment of neuropsychiatric disorders commonly.
Collapse
Affiliation(s)
- Yutaka Nakagawa
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Shizuo Yamada
- Center for Pharma-Food Research (CPFR), Division of Pharmaceutical Sciences, Graduate School of Integrative Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| |
Collapse
|
42
|
Isik S, Yeman Kiyak B, Akbayir R, Seyhali R, Arpaci T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023; 12:cells12071012. [PMID: 37048085 PMCID: PMC10093562 DOI: 10.3390/cells12071012] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson’s Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as “homeostatic” but upon pathological stimuli, they switch to the “reactive state”. Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Uskudar, Istanbul 34662, Turkey
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Correspondence: ; Tel.: +90-216-400-2222 (ext. 2462)
| | - Bercem Yeman Kiyak
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Medicine, Institute of Hamidiye Health Sciences, University of Health Sciences, Uskudar, Istanbul 34668, Turkey
| | - Rumeysa Akbayir
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Rama Seyhali
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Tahire Arpaci
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| |
Collapse
|
43
|
Miao Y, Zhao GL, Cheng S, Wang Z, Yang XL. Activation of retinal glial cells contributes to the degeneration of ganglion cells in experimental glaucoma. Prog Retin Eye Res 2023; 93:101169. [PMID: 36736070 DOI: 10.1016/j.preteyeres.2023.101169] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Elevation of intraocular pressure (IOP) is a major risk factor for neurodegeneration in glaucoma. Glial cells, which play an important role in normal functioning of retinal neurons, are well involved into retinal ganglion cell (RGC) degeneration in experimental glaucoma animal models generated by elevated IOP. In response to elevated IOP, mGluR I is first activated and Kir4.1 channels are subsequently inhibited, which leads to the activation of Müller cells. Müller cell activation is followed by a complex process, including proliferation, release of inflammatory and growth factors (gliosis). Gliosis is further regulated by several factors. Activated Müller cells contribute to RGC degeneration through generating glutamate receptor-mediated excitotoxicity, releasing cytotoxic factors and inducing microglia activation. Elevated IOP activates microglia, and following morphological and functional changes, these cells, as resident immune cells in the retina, show adaptive immune responses, including an enhanced release of pro-inflammatory factors (tumor neurosis factor-α, interleukins, etc.). These ATP and Toll-like receptor-mediated responses are further regulated by heat shock proteins, CD200R, chemokine receptors, and metabotropic purinergic receptors, may aggravate RGC loss. In the optic nerve head, astrogliosis is initiated and regulated by a complex reaction process, including purines, transmitters, chemokines, growth factors and cytokines, which contributes to RGC axon injury through releasing pro-inflammatory factors and changing extracellular matrix in glaucoma. The effects of activated glial cells on RGCs are further modified by the interplay among different types of glial cells. This review is concluded by presenting an in-depth discussion of possible research directions in this field in the future.
Collapse
Affiliation(s)
- Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Guo-Li Zhao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
44
|
Xu J, Zhang L, Li M, He X, Luo J, Wu R, Hong Z, Zheng H, Hu X. TREM2 mediates physical exercise-promoted neural functional recovery in rats with ischemic stroke via microglia-promoted white matter repair. J Neuroinflammation 2023; 20:50. [PMID: 36829205 PMCID: PMC9960657 DOI: 10.1186/s12974-023-02741-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/17/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND The repair of white matter injury is of significant importance for functional recovery after ischemic stroke, and the up-regulation of triggering receptors expressed on myeloid cells 2 (TREM2) after ischemic stroke is neuroprotective and implicated in remyelination. However, the lack of effective therapies calls for the need to investigate the regenerative process of remyelination and the role of rehabilitation therapy. This study sought to investigate whether and how moderate physical exercise (PE) promotes oligodendrogenesis and remyelination in rats with transient middle cerebral artery occlusion (tMCAO). METHODS Male Sprague-Dawley rats (weighing 250-280 g) were subjected to tMCAO. AAV-shRNA was injected into the lateral ventricle to silence the Trem2 gene before the operation. The rats in the physical exercise group started electric running cage training at 48 h after the operation. The Morris water maze and novel object recognition test were used to evaluate cognitive function. Luxol fast blue staining, diffusion tensor imaging, and electron microscopy were used to observe myelin injury and repair. Immunofluorescence staining was applied to observe the proliferation and differentiation of oligodendrocyte precursor cells (OPCs). Expression of key molecules were detected using immunofluorescence staining, quantitative real-time polymerase chain reaction, Western blotting, and Enzyme-linked immunosorbent assay, respectively. RESULTS PE exerted neuroprotective efects by modulating microglial state, promoting remyelination and recovery of neurological function of rats over 35 d after stroke, while silencing Trem2 expression in rats suppressed the aforementioned effects promoted by PE. In addition, by leveraging the activin-A neutralizing antibody, we found a direct beneficial effect of PE on microglia-derived activin-A and its subsequent role on oligodendrocyte differentiation and remyelination mediated by the activin-A/Acvr axis. CONCLUSIONS The present study reveals a novel regenerative role of PE in white matter injury after stroke, which is mediated by upregulation of TREM2 and microglia-derived factor for oligodendrocytes regeneration. PE is an effective therapeutic approach for improving white matter integrity and alleviating neurological function deficits after ischemic stroke.
Collapse
Affiliation(s)
- Jinghui Xu
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Liying Zhang
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Mingyue Li
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Xiaofei He
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Jing Luo
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Rui Wu
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Zhongqiu Hong
- grid.12981.330000 0001 2360 039XDepartment of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
45
|
Lampiasi N, Bonaventura R, Deidda I, Zito F, Russo R. Inflammation and the Potential Implication of Macrophage-Microglia Polarization in Human ASD: An Overview. Int J Mol Sci 2023; 24:2703. [PMID: 36769026 PMCID: PMC9916462 DOI: 10.3390/ijms24032703] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous collection of neurodevelopmental disorders, difficult to diagnose and currently lacking treatment options. The possibility of finding reliable biomarkers useful for early identification would offer the opportunity to intervene with treatment strategies to improve the life quality of ASD patients. To date, there are many recognized risk factors for the development of ASD, both genetic and non-genetic. Although genetic and epigenetic factors may play a critical role, the extent of their contribution to ASD risk is still under study. On the other hand, non-genetic risk factors include pollution, nutrition, infection, psychological states, and lifestyle, all together known as the exposome, which impacts the mother's and fetus's life, especially during pregnancy. Pathogenic and non-pathogenic maternal immune activation (MIA) and autoimmune diseases can cause various alterations in the fetal environment, also contributing to the etiology of ASD in offspring. Activation of monocytes, macrophages, mast cells and microglia and high production of pro-inflammatory cytokines are indeed the cause of neuroinflammation, and the latter is involved in ASD's onset and development. In this review, we focused on non-genetic risk factors, especially on the connection between inflammation, macrophage polarization and ASD syndrome, MIA, and the involvement of microglia.
Collapse
Affiliation(s)
- Nadia Lampiasi
- Istituto per la Ricerca e l’Innovazione Biomedica IRIB, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | | | | | | | | |
Collapse
|
46
|
Mado H, Adamczyk-Sowa M, Sowa P. Role of Microglial Cells in the Pathophysiology of MS: Synergistic or Antagonistic? Int J Mol Sci 2023; 24:ijms24031861. [PMID: 36768183 PMCID: PMC9916250 DOI: 10.3390/ijms24031861] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Many studies indicate an important role of microglia and their cytokines in the pathophysiology of multiple sclerosis (MS). Microglia are the macrophages of the central nervous system (CNS). They have many functions, such as being "controllers" of the CNS homeostasis in pathological and healthy conditions, playing a key role in the active immune defense of the CNS. Macroglia exhibit a dual role, depending on the phenotype they adopt. First, they can exhibit neurotoxic effects, which are harmful in the case of MS. However, they also show neuroprotective and regenerative effects in this disease. Many of the effects of microglia are mediated through the cytokines they secrete, which have either positive or negative properties. Neurotoxic and pro-inflammatory effects can be mediated by microglia via lipopolysaccharide and gamma interferon. On the other hand, the mediators of anti-inflammatory and protective effects secreted by microglia can be, for example, interleukin-4 and -13. Further investigation into the role of microglia in MS pathophysiology may perhaps lead to the discovery of new therapies for MS, as recent research in this area has been very promising.
Collapse
Affiliation(s)
- Hubert Mado
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
- Correspondence: ; Tel.: +48-695948463; Fax: +48-323704597
| | - Monika Adamczyk-Sowa
- Department of Neurology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Paweł Sowa
- Department of Otorhinolaryngology and Oncological Laryngology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| |
Collapse
|
47
|
Guo B, Zhang M, Hao W, Wang Y, Zhang T, Liu C. Neuroinflammation mechanisms of neuromodulation therapies for anxiety and depression. Transl Psychiatry 2023; 13:5. [PMID: 36624089 PMCID: PMC9829236 DOI: 10.1038/s41398-022-02297-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Mood disorders are associated with elevated inflammation, and the reduction of symptoms after multiple treatments is often accompanied by pro-inflammation restoration. A variety of neuromodulation techniques that regulate regional brain activities have been used to treat refractory mood disorders. However, their efficacy varies from person to person and lack reliable indicator. This review summarizes clinical and animal studies on inflammation in neural circuits related to anxiety and depression and the evidence that neuromodulation therapies regulate neuroinflammation in the treatment of neurological diseases. Neuromodulation therapies, including transcranial magnetic stimulation (TMS), transcranial electrical stimulation (TES), electroconvulsive therapy (ECT), photobiomodulation (PBM), transcranial ultrasound stimulation (TUS), deep brain stimulation (DBS), and vagus nerve stimulation (VNS), all have been reported to attenuate neuroinflammation and reduce the release of pro-inflammatory factors, which may be one of the reasons for mood improvement. This review provides a better understanding of the effective mechanism of neuromodulation therapies and indicates that inflammatory biomarkers may serve as a reference for the assessment of pathological conditions and treatment options in anxiety and depression.
Collapse
Affiliation(s)
- Bingqi Guo
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Mengyao Zhang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Wensi Hao
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China
| | - Yuping Wang
- grid.413259.80000 0004 0632 3337Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Neuromodulation, Beijing, 100053 China ,grid.24696.3f0000 0004 0369 153XInstitute of sleep and consciousness disorders, Center of Epilepsy, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069 China
| | - Tingting Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| | - Chunyan Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China. .,Beijing Key Laboratory of Neuromodulation, Beijing, 100053, China.
| |
Collapse
|
48
|
Rau J, Weise L, Moore R, Terminel M, Brakel K, Cunningham R, Bryan J, Stefanov A, Hook MA. Intrathecal minocycline does not block the adverse effects of repeated, intravenous morphine administration on recovery of function after SCI. Exp Neurol 2023; 359:114255. [PMID: 36279935 DOI: 10.1016/j.expneurol.2022.114255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 09/18/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
Opioids are among the most effective analgesics for the management of pain in the acute phase of a spinal cord injury (SCI), and approximately 80% of patients are treated with morphine in the first 24 h following SCI. We have found that morphine treatment in the first 7 days after SCI increases symptoms of pain at 42 days post-injury and undermines the recovery of locomotor function in a rodent model. Prior research has implicated microglia/macrophages in opioid-induced hyperalgesia and the development of neuropathic pain. We hypothesized that glial activation may also underlie the development of morphine-induced pain and cell death after SCI. Supporting this hypothesis, our previous studies found that intrathecal and intravenous morphine increase the number of activated microglia and macrophages present at the spinal lesion site, and that the adverse effects of intrathecal morphine can be blocked with intrathecal minocycline. Recognizing that the cellular expression of opioid receptors, and the intracellular signaling pathways engaged, can change with repeated administration of opioids, the current study tested whether minocycline was also protective with repeated intravenous morphine administration, more closely simulating clinical treatment. Using a rat model of SCI, we co-administered intravenous morphine and intrathecal minocycline for the first 7 days post injury and monitored sensory and locomotor recovery. Contrary to our hypothesis and previous findings with intrathecal morphine, we found that minocycline did not prevent the negative effects of morphine. Surprisingly, we also found that intrathecal minocycline alone is detrimental for locomotor recovery after SCI. Using ex vivo cell cultures, we investigated how minocycline and morphine altered microglia/macrophage function. Commensurate with published studies, we found that minocycline blocked the effects of morphine on the release of pro-inflammatory cytokines but, like morphine, it increased glial phagocytosis. While phagocytosis is critical for the removal of cellular and extracellular debris at the spinal injury site, increased phagocytosis after injury has been linked to the clearance of stressed but viable neurons and protracted inflammation. In sum, our data suggest that both morphine and minocycline alter the acute immune response, and reduce locomotor recovery after SCI.
Collapse
Affiliation(s)
- Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Lara Weise
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA.
| | - Robbie Moore
- Department of Microbial Pathogenesis and Immunology, Texas A&M Institute for Neuroscience, Address: 8447 Riverside Parkway, Medical and Research Education Building 2, Bryan, TX 77807, USA.
| | - Mabel Terminel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA
| | - Kiralyn Brakel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA
| | - Rachel Cunningham
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA
| | - Jessica Bryan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Alexander Stefanov
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| |
Collapse
|
49
|
Gupta DP, Park SH, Lee YS, Lee S, Lim S, Byun J, Cho IH, Song GJ. Daphne genkwa flower extract promotes the neuroprotective effects of microglia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154486. [PMID: 36240609 DOI: 10.1016/j.phymed.2022.154486] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Microglia are innate immune cells in the central nervous system that play a crucial role in neuroprotection by releasing neurotrophic factors, removing pathogens through phagocytosis, and regulating brain homeostasis. The constituents extracted from the roots and stems of the Daphne genkwa plant have shown neuroprotective effects in an animal model of Parkinson's disease. However, the effect of Daphne genkwa plant extract on microglia has yet to be demonstrated. PURPOSE To study the anti-inflammatory and neuroprotective effects of Daphne genkwa flower extract (GFE) in microglia and explore the underlying mechanisms. METHODS In-vitro mRNA expression levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), inducible nitric oxide synthase, Arginase1, and brain derived neurotropic factor (BDNF) were analyzed by reverse transcription polymerase chain reaction in microglia cells. Nitric oxide (NO) and TNF-α protein were respectively analyzed by Griess reagent and Enzyme Linked Immunosorbent Assay. Immunoreactivity of Iba-1, Neu-N, and BDNF in mouse brain were analyzed by immunofluorescence staining. Phagocytosis capacity of microglia was examined using fluorescent zymosan-red particles. RESULTS GFE significantly inhibited lipopolysaccharide (LPS)-induced neuroinflammation and promoted neuroprotection both in vitro and in vivo. First, GFE inhibited the LPS-induced inflammatory factors NO, iNOS, and TNF-α in microglial cell lines and primary glial cells, thus demonstrating anti-inflammatory effects. Arginase1 and BDNF mRNA levels were increased in primary glial cells treated with GFE. Phagocytosis was also increased in microglia treated with GFE, suggesting a neuroprotective effect of GFE. In vivo, neuroprotective and anti-neuroinflammatory effects of GFE were also found in the mouse brain, as oral administration of GFE significantly inhibited LPS-induced neuronal loss and inflammatory activation of microglia. CONCLUSION GFE has anti-inflammatory effects and promotes microglial neuroprotective effects. GFE inhibited the pro-inflammatory mediators and enhanced neuroprotective microglia activity by increasing BDNF expression and phagocytosis. These novel findings of the GFE effect on microglia show an innovative approach that can potentially promote neuroprotection for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Deepak Prasad Gupta
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea
| | - Sung Hee Park
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea
| | - Young-Sun Lee
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea; The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong, Korea
| | - Sujin Lim
- Department of Life Science, The Catholic University of Korea, Bucheon, Korea
| | - Jiin Byun
- Department of Life Science, The Catholic University of Korea, Bucheon, Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Korea.
| | - Gyun Jee Song
- Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung, Gangwon-do, Korea; The Convergence Institute of Healthcare and Medical Science, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Korea.
| |
Collapse
|
50
|
Gao S, Jiang Y, Chen Z, Zhao X, Gu J, Wu H, Liao Y, Sun H, Wang J, Chen W. Metabolic Reprogramming of Microglia in Sepsis-Associated Encephalopathy: Insights from Neuroinflammation. Curr Neuropharmacol 2023; 21:1992-2005. [PMID: 36529923 PMCID: PMC10514522 DOI: 10.2174/1570159x21666221216162606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction caused by sepsis that manifests as a range of brain dysfunctions from delirium to coma. It is a relatively common complication of sepsis associated with poor patient prognosis and mortality. The pathogenesis of SAE involves neuroinflammatory responses, neurotransmitter dysfunction, blood-brain barrier (BBB) disruption, abnormal blood flow regulation, etc. Neuroinflammation caused by hyperactivation of microglia is considered to be a key factor in disease development, which can cause a series of chain reactions, including BBB disruption and oxidative stress. Metabolic reprogramming has been found to play a central role in microglial activation and executive functions. In this review, we describe the pivotal role of energy metabolism in microglial activation and functional execution and demonstrate that the regulation of microglial metabolic reprogramming might be crucial in the development of clinical therapeutics for neuroinflammatory diseases like SAE.
Collapse
Affiliation(s)
- Shenjia Gao
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yi Jiang
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhaoyuan Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Jiahui Gu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Han Wu
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yun Liao
- Shanghai Medical College of Fudan University, Shanghai, China
| | - Hao Sun
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jun Wang
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Wankun Chen
- Department of Anesthesiology, Cancer Center, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Fudan Zhangjiang Institute, Shanghai, 201203, China
| |
Collapse
|