1
|
Chung E, Zhang D, Gonzalez Porras M, Hsu CG. TREM2 as a regulator of obesity-induced cardiac remodeling: mechanisms and therapeutic insights. Am J Physiol Heart Circ Physiol 2025; 328:H1073-H1082. [PMID: 40152357 DOI: 10.1152/ajpheart.00075.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/17/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are global health challenges that significantly increase the risk of cardiovascular diseases (CVD). Advances in immunometabolism have identified triggering receptor expressed on myeloid cells 2 (TREM2) as a key regulator of macrophage function, lipid metabolism, and inflammation resolution. Although extensively studied in neurodegenerative diseases, TREM2's role in metabolic disorders and cardiovascular health is an emerging area of research. This review explores TREM2's molecular structure and functions, emphasizing its contributions to immunometabolic regulation in obesity and T2DM. Evidence from preclinical models demonstrates that TREM2 modulates macrophage-driven inflammatory responses, lipid clearance, plaque stability, fibrosis, and myocardial remodeling. Translational findings suggest that TREM2 expression correlates with cardiometabolic outcomes, underscoring its potential as a therapeutic target. Key knowledge gaps include TREM2's temporal dynamics during disease progression, sex-specific effects, and interactions with recruited or resident macrophage activation in obesity and T2DM. Integrating mechanistic and translational insights is critical to harness TREM2's immunoregulatory potential for improving CVD outcomes in metabolic disorders.
Collapse
Affiliation(s)
- Eunhee Chung
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| | - David Zhang
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Maria Gonzalez Porras
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Chia George Hsu
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| |
Collapse
|
2
|
Li HD, Zheng JY, Tan KW, Su JX, Chen W, Pang RK, Wu GL, Qiu YH, Li XX, Cai YF, Zhang SJ. Salvianolic acid B (SalB) improves high-fat diet (HFD)-caused cognitive impairment in mice by modulating the Trem2/Dap12 pathway in vivo and in vitro. Int Immunopharmacol 2025; 153:114461. [PMID: 40101423 DOI: 10.1016/j.intimp.2025.114461] [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: 12/13/2024] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025]
Abstract
Salvianolic acid B (SalB), which extracted from Salvia miltiorrhiza Bunge (Labiatae), is a traditional Chinese medicine. SalB is widely used in nervous system diseases. This study evaluated the protective effect of SalB on high-fat diet (HFD)-induced cognitive impairment and its mechanisms in vivo and in vitro. The behavior tests demonstrated that SalB alleviated motor skills and learning capacity in HFD mice. Animal experiments have confirmed that SalB reduced the mRNA expression of inflammatory markers and the Trem2/Dap12 pathway in HIP. Furthermore, SalB inhibited the microglia Trem2/Dap12 pathway in HIP. In vivo, palmitic acid (PA) was used to intervene in BV2 cells to construct an inflammatory. SalB reduced the mRNA expression of inflammatory markers and inhibited the Trem2/Dap12 pathway in BV2 cells. In conclusion, SalB treatment may serve as a possible therapy for cognitive impairment induced by HFD.
Collapse
Affiliation(s)
- Hong-Dan Li
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Kai-Wen Tan
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Jin-Xun Su
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Wei Chen
- Department of Neurology, Nanning Hospital of Traditional Chinese Medicine, Nanning 530000, China
| | - Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Guang-Liang Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Yu-Hui Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China
| | - Xiao-Xiao Li
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong 999077, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China.
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China; Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou 510000, China.
| |
Collapse
|
3
|
Islam R, Choudhary HH, Zhang F, Mehta H, Yoshida J, Thomas AJ, Hanafy K. Microglial TLR4-Lyn kinase is a critical regulator of neuroinflammation, Aβ phagocytosis, neuronal damage, and cell survival in Alzheimer's disease. Sci Rep 2025; 15:11368. [PMID: 40175501 PMCID: PMC11965285 DOI: 10.1038/s41598-025-96456-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: 01/12/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
Disease-Associated Microglia (DAM) are a focus in Alzheimer's disease (AD) research due to their central involvement in the response to amyloid-beta plaques. Microglial Toll-like receptor 4 (TLR4) is instrumental in the binding of fibrillary amyloid proteins, while Lyn kinase (Lyn) is a member of the Src family of non-receptor tyrosine kinases involved in immune signaling. Lyn is a novel, non-canonical, intracellular adaptor with diverse roles in cell-specific signaling which directly binds to TLR4 to modify its function. Lyn can be activated in response to TLR4 stimulation, leading to phosphorylation of various substrates and modulation of inflammatory and phagocytosis signaling pathways. Here, we investigated the TLR4-Lyn interaction in neuroinflammation using WT, 5XFAD, and 5XFAD x Lyn-/- mouse models by western blotting (WB), co-immunoprecipitation (co-IP), immunohistochemistry (IHC) and flow cytometric (FC) analysis. A spatial transcriptomic analysis of microglia in WT, 5XFAD, and 5XFAD x Lyn-/- mice revealed essential genes involved in neuroinflammation, Aβ phagocytosis, and neuronal damage. Finally, we explored the effects of a synthetic, TLR4-Lyn modulator protein (TLIM) through an in vitro AD model using primary murine microglia. Our WB, co-IP, IHC, and FC data show an increased, novel, direct protein-protein interaction between TLR4 and Lyn kinase in the brains of 5XFAD mice compared to WT. Furthermore, in the absence of Lyn (5XFAD x Lyn-/- mice); increased expression of protective Syk kinase was observed, enhanced microglial Aβ phagocytosis, increased astrocyte activity, decreased neuronal dystrophy, and a further increase in the cell survival signaling and protective DAM population was noted. The DAM population in 5XFAD mice which produce more inflammatory cytokines and phagocytose more Aβ were observed to express greater levels of TLR4 and Lyn. Pathway analysis comparison between WT, 5XFAD, and 5XFAD x Lyn-/- mice supported these findings via our microglial spatial transcriptomic analysis. Finally, we created an in vitro co-culture system with primary murine microglial and primary murine hippocampal cells exposed to Aβ as a model of AD. When these co-cultures were treated with our TLR4-Lyn Interaction Modulators (TLIMs), an increase in Aβ phagocytosis and a decrease in neuronal dystrophy was seen. Lyn kinase has a central role in modulating TLR4-induced inflammation and Syk-induced protection in a 5XFAD mouse model. Our TLIMs ameliorate AD sequalae in an in vitro model of AD and could be a promising therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Feng Zhang
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Jun Yoshida
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Ajith J Thomas
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurosurgery, Cooper University Health Care, Camden, NJ, USA
| | - Khalid Hanafy
- Cooper Medical School of Rowan University, Camden, NJ, USA.
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School of Rowan University, 401 Broadway, Camden, NJ, 08103, USA.
| |
Collapse
|
4
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
5
|
Que X, Zhang T, Liu X, Yin Y, Xia X, Gong P, Song W, Qin Q, Xu ZQD, Tang Y. The role of TREM2 in myelin sheath dynamics: A comprehensive perspective from physiology to pathology. Prog Neurobiol 2025; 247:102732. [PMID: 40021075 DOI: 10.1016/j.pneurobio.2025.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Demyelinating disorders, characterizing by the loss of myelin integrity, present significant challenges due to their impact on neurological function and lack of effective treatments. Understanding the mechanisms underlying myelin damage is crucial for developing therapeutic strategies. Triggering receptor expressed on myeloid cells 2 (TREM2), a pivotal immune receptor predominantly found on microglial cells, plays essential roles in phagocytosis and lipid metabolism, vital processes in neuroinflammation and immune regulation. Emerging evidence indicates a close relationship between TREM2 and various aspects of myelin sheath dynamics, including maintenance, response to damage, and regeneration. This review provides a comprehensive discussion of TREM2's influence on myelin physiology and pathology, highlighting its therapeutic potential and putative mechanisms in the progression of demyelinating disorders.
Collapse
Affiliation(s)
- Xinwei Que
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China; Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Tongtong Zhang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Xueyu Liu
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Yunsi Yin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Xinyi Xia
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China
| | - Ping Gong
- Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Weiyi Song
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China; Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China
| | - Qi Qin
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China.
| | - Zhi-Qing David Xu
- Departments of Neurobiology and Pathology, Capital Medical University, Beijing 100069, China.
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing 100053, China; Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing 100053, China.
| |
Collapse
|
6
|
Li D, Pan L, Chen M, Zhang X, Jiang Z. TREM2 protects against LPS-induced murine acute lung injury through suppressing macrophage ferroptosis. Int Immunopharmacol 2025; 150:114247. [PMID: 39946766 DOI: 10.1016/j.intimp.2025.114247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/03/2025]
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) is a cell surface receptor, majorly expressed by granulocytes, monocytes and macrophages. We in this study showed that TREM2 was downregulated in the lipopolysaccharide (LPS)-treated macrophages and murine acute lung injury (ALI) through activation of p38 MAPK and STAT6 signaling. Over-expression of TREM2 reduced the expression of DNAX-activation protein 12 (DAP12), pro-inflammatory cytokines (TNF-α, IL-6, IL-1β), Malondialdehyde (MDA) and hemosiderin accumulation in LPS-treated macrophages. Knockdown of TREM2 expression elevated the expression of IL-6, reactive oxygen species (ROS), lactate dehydrogenases (LDH) and hemosiderin accumulation. Intratracheal adoptive transfer of TREM2-overexpressing macrophages effectively suppressed the lung inflammation and pro-inflammatory cytokine expression in murine ALI. While downregulation of TREM2 enhanced the lung inflammation in the lung tissues of murine ALI. Therefore, TREM2/DAP12 axis is involved in macrophage ferroptosis and attenuation of murine ALI. TREM2 would be a novel therapeutic target in murine ALI and patients with acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Dandan Li
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Linyue Pan
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mengjie Chen
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaoju Zhang
- Department of Pulmonary and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Chen K, Li F, Zhang S, Chen Y, Ikezu TC, Li Z, Martens YA, Qiao W, Meneses A, Zhu Y, Xhafkollari G, Bu G, Zhao N. Enhancing TREM2 expression activates microglia and modestly mitigates tau pathology and neurodegeneration. J Neuroinflammation 2025; 22:93. [PMID: 40122810 PMCID: PMC11931752 DOI: 10.1186/s12974-025-03420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
TREM2, a microglia-specific receptor, is strongly associated with Alzheimer's disease (AD) risk, mediating microglial responses to amyloid pathology critical to AD development. However, its role in tau pathology and neurodegeneration remains unclear. Using the PS19 tauopathy mouse model with inducible overexpression of human wild-type TREM2 (TREM2-WT) or the R47H variant (TREM2-R47H), we show that increasing TREM2-WT expression modestly reduces soluble phosphorylated tau levels and mildly preserves neuronal integrity. Single-cell RNA sequencing reveals that TREM2-WT robustly enhances microglial activation, characterized by a disease-associated microglia (DAM) signature. In contrast, TREM2-R47H overexpression exhibits a loss-of-function phenotype, with no significant impact on tau levels, neurodegeneration, or microglial activation. These findings highlight the role of TREM2 in modulating microglial activity and its influence on tau pathology and neurodegeneration, providing important insights for the future development of therapies targeting TREM2 or microglial pathways in AD or other tauopathies.
Collapse
Affiliation(s)
- Kai Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Fuyao Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Shuwen Zhang
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Tadafumi C Ikezu
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Axel Meneses
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Gisela Xhafkollari
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
- Clinical and Translational Science Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
8
|
Wang Q, Guo C, Wang T, Shuai P, Wu W, Huang S, Li Y, Zhao P, Zeng C, Yi L. Drug protection against radiation-induced neurological injury: mechanisms and developments. Arch Toxicol 2025; 99:851-863. [PMID: 39724149 DOI: 10.1007/s00204-024-03933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
In daily life, individuals are frequently exposed to various forms of radiation, which, when adhering to safety standards, typically result in relatively minor health effects. However, accidental exposure to radiation levels that exceed these safety standards can lead to significant health consequences. This study focuses on the analysis of radiation-induced damage to the nervous system and the mechanisms of pharmacological protection. The findings indicate that radiation can adversely affect neural structures, memory, and neurobehaviour. A range of pharmacological agents, including traditional Chinese medicine, Western medicine, and other therapeutic drugs, can be employed to safeguard the nervous system from radiation damage. The primary protective mechanisms of these agents encompass antioxidant effects, attenuation of apoptosis, and reduction of neurogenesis. A comprehensive review of these topics will offer new insights for the development and investigation of drugs aimed at mitigating radiation-induced damage to the nervous system.
Collapse
Affiliation(s)
- Qingyu Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Caimao Guo
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tiantian Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Peimeng Shuai
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenyu Wu
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shuqi Huang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuanyuan Li
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pei Zhao
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengkai Zeng
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Lan Yi
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
9
|
Li L, Xu N, He Y, Tang M, Yang B, Du J, Chen L, Mao X, Song B, Hua Z, Tang B, Lee SMY. Dehydroervatamine as a promising novel TREM2 agonist, attenuates neuroinflammation. Neurotherapeutics 2025; 22:e00479. [PMID: 39609160 PMCID: PMC12014313 DOI: 10.1016/j.neurot.2024.e00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
Microglia play a dual role in neuroinflammatory disorders that affect millions of people worldwide. These specialized cells are responsible for the critical clearance of debris and toxic proteins through endocytosis. However, activated microglia can secrete pro-inflammatory mediators, potentially exacerbating neuroinflammation and harming adjacent neurons. TREM2, a cell surface receptor expressed by microglia, is implicated in the modulation of neuroinflammatory responses. In this study, we investigated if and how Dehydroervatamine (DHE), a natural alkaloid, reduced the inflammatory phenotype of microglia and suppressed neuroinflammation. Our findings revealed that DHE was directly bound to and activated TREM2. Moreover, DHE effectively suppressed the production of pro-inflammatory cytokines, restored mitochondrial function, and inhibited NLRP3 inflammasome activation via activating the TREM2/DAP12 signaling pathway in LPS-stimulated BV2 microglial cells. Notably, silencing TREM2 abolished the suppression effect of DHE on the neuroinflammatory response, mitochondrial dysfunction, and NF-κB/NLRP3 pathways in vitro. Additionally, DHE pretreatment exhibited remarkable neuroprotective effects, as evidenced by increased neuronal viability and reduced apoptotic cell numbers in SH-SY5Y neuroblastoma cells co-cultured with LPS-stimulated BV2 microglia. Furthermore, in our zebrafish model, DHE pretreatment effectively alleviated behavioral impairments, reduced neutrophil aggregation, and suppressed neuroinflammation in the brain by regulating TREM2/NF-κB/NLRP3 pathways after intraventricular LPS injection. These findings provide novel insights into the potent protective effects of DHE as a promising novel TREM2 agonist against LPS-induced neuroinflammation, revealing its potential therapeutic role in the treatment of central nervous system diseases associated with neuroinflammation.
Collapse
Affiliation(s)
- Lin Li
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Nan Xu
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yulin He
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Mingsui Tang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Binrui Yang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co, Ltd, Shanghai, China
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhou Hua
- Chinese Medicine Guangdong Laboratory (Hengqin Laboratory), Guangdong-Macao ln-Depth Cooperation Zone in Hengqin, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Benqin Tang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; PolyU-BGI Joint Research Centre for Genomics and Synthetic Biology in Global Ocean Resource, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
10
|
Ayinla MT, Asuku AO. The neurotoxic effects of lead acetate and the abrogating actions of 6-gingerol-rich extract of ginger via modulation of antioxidant defence system, pro-inflammatory markers, and apoptotic cascade. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03873-x. [PMID: 39937255 DOI: 10.1007/s00210-025-03873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/01/2025] [Indexed: 02/13/2025]
Abstract
Lead exposure is a public health concern and it has been linked to cognitive deficit, memory impairment, and neurotoxicity. This study was designed to investigate the effect of 6-gingerol-rich extract of ginger (6-GREG) on oxidative stress, inflammation, and apoptosis in lead acetate (PbAc)-induced neurotoxicity in male Wistar rats. Twenty-five (25) male Wistar rats in total were divided into five groups at random (n = 5). The control group received 0.5 ml of normal saline, the PbAc-treated group received 7.5 mg/kg of PbAc, the vitamin C, 6-GREG (100), and 6-GREG (200) groups received 7.5 mg/kg of PbAc followed by administration of vitamin C (100 mg/kg), 6-GREG (100 mg/kg), and 6-GREG (200 mg/kg) respectively for 2 weeks. Following behavioral tests, the rats were euthanized, and their brain tissues were homogenized for biochemical analysis. When compared to the control group, the administration of PbAc caused behavioral alterations as well as a significant (p < 0.05) decrease in the activities of catalase, glutathione peroxidase, as well as reduced glutathione and Bcl-2 levels in the PbAc-treated group. Furthermore, the PbAc-treated group showed a statistically significant rise (p < 0.05) in brain acetylcholinesterase, malondialdehyde, nitric oxide, interleukin-1-beta, tumor necrosis factor-α, and caspase-9 levels in comparison to the control. Administration of both 100 mg/kg and 200 mg/kg of 6-GREG effectively reversed these behavioral and biochemical changes in 6-GREG (100)- and 6-GREG (200)-treated groups respectively compared to the PbAc-treated group. Consequently, the study reveals the role of 6-GREG in attenuating PbAc-induced neurotoxicity and brain damage via antioxidative, anti-inflammatory, and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Maryam Tayo Ayinla
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Ilorin, Nigeria
| | - Abraham Olufemi Asuku
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Ilorin, Nigeria.
- Bioresources Development Centre, National Biotechnology Research and Development Agency (NBRDA), Ogbomoso, Nigeria.
| |
Collapse
|
11
|
Farzan M, Saberi-Rounkian M, Asadi-Rizi A, Heidari Z, Farzan M, Fathi M, Aghaei A, Azadegan-Dehkordi F, Bagheri N. The emerging role of the microglia triggering receptor expressed on myeloid cells (TREM) 2 in multiple sclerosis. Exp Neurol 2025; 384:115071. [PMID: 39586397 DOI: 10.1016/j.expneurol.2024.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The chronic inflammatory condition known as multiple sclerosis (MS) causes inflammation and demyelination in the central nervous system (CNS). The activation of multiple cell types, including the CNS's resident immune cells called microglia, is a component of the immunological response in MS. Recently, the triggering receptor expressed on myeloid cells (TREM) family has emerged as a crucial player in modulating microglial function and subsequent neuroinflammation. Understanding the role of TREM receptors in MS pathogenesis could provide insightful information on how to develop new therapeutic approaches. MAIN BODY The TREM family consists of several receptors, including TREM-1 and TREM-2, which can be expressed on both immune cells, such as myeloid cells and microglia, and non-immune cells. These receptors interact with their respective ligands and regulate signaling pathways, ultimately leading to the control of microglial activation and inflammatory reactions. TREM-2, in particular, has garnered significant interest because of its connection with MS and other neurodegenerative diseases. The activation of microglia through TREM receptors in MS is thought to influence the equilibrium between helpful and detrimental inflammatory responses. TREM receptors can promote the phagocytosis of myelin debris and remove apoptotic cells, thus contributing to tissue repair and regeneration. However, excessive or dysregulated activation of microglia mediated by TREM receptors can lead to the release of pro-inflammatory cytokines and neurotoxic factors, exacerbating neuroinflammation and neurodegeneration in MS. CONCLUSION The emerging role of the TREM family in demyelinating diseases highlights the importance of microglia in disease pathogenesis. Understanding the mechanisms by which TREM receptors modulate microglial function can provide valuable insights into the development of targeted therapies for these disorders. By selectively targeting TREM receptors, it may be possible to harness their beneficial effects on tissue repair while dampening their detrimental pro-inflammatory responses. Further research is warranted to elucidate the precise signaling pathways and ligand interactions involved in TREM-mediated microglial activation, which could uncover novel therapeutic avenues for treating MS and other neuroinflammatory disorders.
Collapse
Affiliation(s)
- Mahan Farzan
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Masoumeh Saberi-Rounkian
- Student Research committee, School of Paramedicine, Guilan University of Medical sciences, Rasht, Iran
| | - Atefeh Asadi-Rizi
- Young researchers and Elite club, Flavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Zahra Heidari
- Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Mahour Farzan
- Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ava Aghaei
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; Medical Plants Research Center, Basic Health Sciences institute, Shahrekord University of Medical sciences, Shahrekord, Iran
| | - Fatemeh Azadegan-Dehkordi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Nader Bagheri
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
12
|
Larson KC, Gergits FW, Renoux AJ, Weisman EJ, Dejanovic B, Huang L, Pandya B, McLaren DG, Lynch BA, Fisher R, Thackaberry E, Gray D, Gaudreault F, Mirescu C. Rescue of in vitro models of CSF1R-related adult-onset leukodystrophy by iluzanebart: mechanisms and therapeutic implications of TREM2 agonism. J Neuroinflammation 2025; 22:26. [PMID: 39891235 PMCID: PMC11783791 DOI: 10.1186/s12974-025-03346-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/03/2025] Open
Abstract
Microglia dysfunction is implicated in several neurodegenerative disorders, including a rare microgliopathy; CSF1R-related adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (CSF1R-ALSP). CSF1R-ALSP is caused by heterozygous loss-of-function mutations in the colony stimulating factor 1 receptor (CSF1R) gene, which encodes a receptor required for the differentiation of myeloid cells, as well as for microglial survival and proliferation. Similar functions have also been ascribed to triggering receptor expressed on myeloid cells 2 (TREM2), which shares an analogous microglia enrichment profile and converging intracellular signaling pathway mediated by spleen associated tyrosine kinase (SYK) and phosphoinositide-3-kinase (PI3K). Iluzanebart is a human monoclonal IgG1, human TREM2 (hTREM2) agonist antibody under development for the treatment of CSF1R-ALSP. To explore the therapeutic hypothesis that loss of CSF1R signaling and related microglial hypofunction can be circumvented via activation of TREM2, we evaluated the potential of iluzanebart to compensate for CSF1R loss-of-function. Herein, we demonstrate that iluzanebart is a potent, dose-dependent, and specific activator of TREM2 signaling in human primary cells. Iluzanebart treatment rescued viability of human monocyte-derived macrophages (hMDM) and induced pluripotent stem cell-derived human microglia (iMGL) in multiple in vitro models of CSF1R-ALSP, including in induced pluripotent stem cell (iPSC) differentiated microglia carrying the heterozygous I794T mutation found in CSF1R-ALSP patients. Additionally, iluzanebart treatment in microglia modulated surface levels of CSF1R, resulting in increased receptor activation as measured by phosphorylation of CSF1R. Differentially expressed genes identified in the hippocampus of mice treated with iluzanebart were exemplary of TREM2 activation and were related to cell proliferation, regulation of inflammatory processes, and innate immune response pathways. Proliferation of microglia, changes in protein levels of specific chemokines identified by gene expression analysis, and increased CSF1R levels were also confirmed in vivo. These findings demonstrate that iluzanebart is a potent and selective TREM2 agonistic antibody, with pharmacology that supports the hypothesis that TREM2 activation can compensate for CSF1R dysfunction and its continued clinical development for individuals with CSF1R-ALSP.
Collapse
Affiliation(s)
- Kelley C Larson
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA.
| | | | - Abigail J Renoux
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | | | | | - Liyue Huang
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | - Bhaumik Pandya
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | - Donald G McLaren
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | - Berkley A Lynch
- Vigil Neuroscience Inc. (Ret), 100 Forge Road, Watertown, MA, 02472, USA
| | - Richard Fisher
- Eikonizo Therapeutics, Inc, 245 Main St, Cambridge, MA, 02142, USA
| | - Evan Thackaberry
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | - David Gray
- Vigil Neuroscience Inc, 100 Forge Road, Watertown, MA, 02472, USA
| | | | | |
Collapse
|
13
|
Fremuth LE, Hu H, van de Vlekkert D, Annunziata I, Weesner JA, Alessandra d'Azzo. Neuraminidase 1 regulates neuropathogenesis by governing the cellular state of microglia via modulation of Trem2 sialylation. Cell Rep 2025; 44:115204. [PMID: 39817909 PMCID: PMC11874873 DOI: 10.1016/j.celrep.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/08/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Neuraminidase 1 (NEU1) cleaves terminal sialic acids from sialoglycoproteins in endolysosomes and at the plasma membrane. As such, NEU1 regulates immune cells, primarily those of the monocytic lineage. Here, we examine how Neu1 influences microglia by modulating the sialylation of full-length Trem2 (Trem2-FL), a multifunctional receptor that regulates microglial survival, phagocytosis, and cytokine production. When Neu1 is deficient/downregulated, Trem2-FL remains sialylated, accumulates intracellularly, and is excessively cleaved into a C-terminal fragment (Trem2-CTF) and an extracellular soluble domain (sTrem2), enhancing their signaling capacities. Sialylated Trem2-FL (Sia-Trem2-FL) does not hinder Trem2-FL-DAP12-Syk complex assembly but impairs signal transduction through Syk, ultimately abolishing Trem2-dependent phagocytosis. Concurrently, Trem2-CTF-DAP12 complexes dampen NF-κB signaling, while sTrem2 propagates Akt-dependent cell survival and NFAT1-mediated production of TNF-α and CCL3. Because NEU1 and Trem2 are implicated in neurodegenerative/neuroinflammatory diseases, including Alzheimer disease and sialidosis, modulating NEU1 activity represents a therapeutic approach to broadly regulate microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Leigh Ellen Fremuth
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Huimin Hu
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Ida Annunziata
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Compliance Office, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jason Andrew Weesner
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
14
|
Bosco DB, Kremen V, Haruwaka K, Zhao S, Wang L, Ebner BA, Zheng J, Xie M, Dheer A, Perry JF, Barath A, Nguyen AT, Worrell GA, Wu LJ. Microglial TREM2 promotes phagocytic clearance of damaged neurons after status epilepticus. Brain Behav Immun 2025; 123:540-555. [PMID: 39353548 PMCID: PMC11924143 DOI: 10.1016/j.bbi.2024.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/15/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024] Open
Abstract
In the central nervous system, triggering receptor expressed on myeloid cells 2 (TREM2) is exclusively expressed by microglia and is critical for microglial proliferation, migration, and phagocytosis. Microglial TREM2 plays an important role in neurodegenerative diseases, such as Alzheimer's disease and amyotrophic lateral sclerosis. However, little is known about how TREM2 affects microglial function within epileptogenesis. To investigate this, we utilized male TREM2 knockout (KO) mice within the intra-amygdala kainic acid seizure model. Electroencephalographic analysis, immunocytochemistry, and RNA sequencing revealed that TREM2 deficiency significantly promoted seizure-induced pathology. We found that TREM2 KO increased both the severity of acute status epilepticus and the number of spontaneous recurrent seizures characteristic of chronic focal epilepsy. Phagocytic clearance of damaged neurons by microglia was also impaired by TREM2 KO and reduced phagocytic activity correlated with increased spontaneous seizures. Analysis of human tissue from patients who underwent surgical resection for drug resistant temporal lobe epilepsy also showed a negative correlation between expression of the microglial phagocytic marker CD68 and focal to bilateral tonic-clonic generalized seizure history. These results indicate that microglial TREM2 and phagocytic activity are important to epileptogenic pathology.
Collapse
MESH Headings
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Animals
- Status Epilepticus/metabolism
- Status Epilepticus/genetics
- Microglia/metabolism
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/genetics
- Mice, Knockout
- Male
- Phagocytosis/physiology
- Phagocytosis/genetics
- Mice
- Neurons/metabolism
- Humans
- Disease Models, Animal
- Kainic Acid
- Mice, Inbred C57BL
- Epilepsy, Temporal Lobe/metabolism
- Epilepsy, Temporal Lobe/genetics
- Seizures/metabolism
- Seizures/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
Collapse
Affiliation(s)
- Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Vaclav Kremen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Koichiro Haruwaka
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lingxiao Wang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Blake A Ebner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jiaying Zheng
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Aastha Dheer
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jadyn F Perry
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Abhijeet Barath
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
15
|
Pashaei S, Shabani S, Mohammadi S, Morozova-Roche LA, Salari N, Rahimi Z, Khodarahmi R. Differential Expression of Neurodegeneration-Related Genes in SH-SY5Y Neuroblastoma Cells Under the Influence of Cyclophilin A: Could the Enzyme be a Likely Trigger and Therapeutic Target for Alzheimer's Disease? Neurochem Res 2024; 50:47. [PMID: 39636462 DOI: 10.1007/s11064-024-04253-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The function and mechanism of Cyclophilin A (CypA) in modulating gene expression associated with Alzheimer's disease (AD) remain unclear. This multifunctional protein is found to be elevated in the cerebrospinal fluid (CSF) of individuals at risk for AD. The cytotoxic effects of CypA, including both wild-type and the mutant R55A, were assessed using the MTT assay. Prior to this evaluation, the purified recombinant protein was validated through enzymatic activity assays and western blot analysis. Following treatment with CypA and transient transfection using the CypA construct, real-time PCR (qRT-PCR) and western blotting were conducted to analyze the expression of factors involved in various signaling pathways, with an emphasis on inflammation, cell death, and intercellular communication. The findings indicate that CypA has a significant impact on the gene expression of factors associated with inflammation and the progression of AD in SH-SY5Y cells. It can be concluded that CypA is capable of regulating gene expression in SH-SY5Y cells, either in a manner dependent on or independent of its enzymatic activity. Additionally, the influence of this multifunctional protein on gene expression is contingent upon the specific site of action, as well as the dosage and duration of exposure to the cells.
Collapse
Affiliation(s)
- Somayeh Pashaei
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sasan Shabani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Nader Salari
- Department of Biostatics, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
16
|
Najdaghi S, Davani DN, Fouladseresht H, Ebrahimi N, Sullman MJM, Moradi M, Eskandari N. The Role of Extracellular Vesicles and Microparticles in Central Nervous System Disorders: Mechanisms, Biomarkers, and Therapeutic Potential. Cell Mol Neurobiol 2024; 44:82. [PMID: 39625540 PMCID: PMC11614997 DOI: 10.1007/s10571-024-01518-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024]
Abstract
Microscopic, membranous vesicles known as extracellular vesicles (EVs) have been proposed to play a role in the mechanisms underlying central nervous system (CNS) diseases. EVs are secreted by a variety of cells, including myeloid, endothelial, microglial, oligodendroglial, and mesenchymal stem cells (MSCs). Body fluids such as plasma, urine, and cerebrospinal fluid (CSF) contain microparticles (MPs). The detection of MPs in CSF may indicate genetic or environmental susceptibility to conditions such as schizophrenia, schizoaffective disorder, and bipolar disorder. MPs of different origins can exhibit changes in specific biomarkers at various stages of the disease, aiding in the diagnosis and monitoring of neurological conditions. However, understanding the role and clinical applications of MPs is complicated by challenges such as their isolation and dual roles within the CNS. In this review, we discuss the history, characteristics, and roles of MPs in CNS diseases. We also provide practical insights for future research and highlight the challenges that obscure the therapeutic potential of MPs.
Collapse
Affiliation(s)
- Soroush Najdaghi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hamed Fouladseresht
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Narges Ebrahimi
- Neuroscience Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Social Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, School of Humanities and Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Marjan Moradi
- Departement of Genetics, School of Science, Shahrekord University, Shahrakord, Iran
| | - Nahid Eskandari
- Immunology Department, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran.
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
17
|
Wang Y, Niu W, Zhu S, Sun J, Lv J, Wang N, Zhang H, Zhang Z, Wang M, Cao L, Li S, Zhai Q, Ma L. STING Agonist cGAMP Attenuates Sleep Deprivation-Induced Neuroinflammation and Cognitive Deficits via TREM2 Up-Regulation. Inflammation 2024; 47:2129-2144. [PMID: 38668837 DOI: 10.1007/s10753-024-02029-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 11/30/2024]
Abstract
Sleep deprivation (SD) has been associated with several adverse effects, including cognitive deficit. Emerging evidence suggests microglia-associated neuroinflammation is a potential trigger of cognitive deficit after SD. Stimulator of interferon genes (STING) constitutes an important factor in host immune response to pathogenic organisms and is found in multiple cells, including microglia. STING is involved in neuroinflammation during neuronal degeneration, although how STING signaling affects SD-induced neuroinflammation remains unexplored. In the present study, the chronic sleep restriction (CSR) model was applied to examine the effects of STING signaling on cognition. The results revealed that cGAMP, a high-affinity and selective STING agonist, significantly improved cognitive deficit, alleviated neural injury, and relieved neuroinflammation in CSR mice by activating the STING-TBK1-IRF3 pathway. Moreover, triggering receptor expressed on myeloid cells 2 (TREM2) was upregulated in CSR mice treated with cGAMP, and this effect was abolished by STING knockout. TREM2 upregulation induced by cGAMP regulated the microglia from pro-inflammatory state to anti-inflammatory state, thereby relieving neuroinflammation in CSR mice. These findings indicate cGAMP-induced STING signaling activation alleviates SD-associated neuroinflammation and cognitive deficit by upregulating TREM2, providing a novel approach for the treatment of SD-related nerve injury.
Collapse
Affiliation(s)
- Yue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Wen Niu
- Department of Physiology and Pathophysiology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shan Zhu
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jianyu Sun
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jianrui Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Ning Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Huijuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zhenni Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Meijuan Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Lingli Cao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shuwei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Qian Zhai
- Department of Anesthesiology and Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Lei Ma
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
18
|
Western D, Timsina J, Wang L, Wang C, Yang C, Phillips B, Wang Y, Liu M, Ali M, Beric A, Gorijala P, Kohlfeld P, Budde J, Levey AI, Morris JC, Perrin RJ, Ruiz A, Marquié M, Boada M, de Rojas I, Rutledge J, Oh H, Wilson EN, Le Guen Y, Reus LM, Tijms B, Visser PJ, van der Lee SJ, Pijnenburg YAL, Teunissen CE, Del Campo Milan M, Alvarez I, Aguilar M, Greicius MD, Pastor P, Pulford DJ, Ibanez L, Wyss-Coray T, Sung YJ, Cruchaga C. Proteogenomic analysis of human cerebrospinal fluid identifies neurologically relevant regulation and implicates causal proteins for Alzheimer's disease. Nat Genet 2024; 56:2672-2684. [PMID: 39528825 PMCID: PMC11831731 DOI: 10.1038/s41588-024-01972-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The integration of quantitative trait loci (QTLs) with disease genome-wide association studies (GWASs) has proven successful in prioritizing candidate genes at disease-associated loci. QTL mapping has been focused on multi-tissue expression QTLs or plasma protein QTLs (pQTLs). We generated a cerebrospinal fluid (CSF) pQTL atlas by measuring 6,361 proteins in 3,506 samples. We identified 3,885 associations for 1,883 proteins, including 2,885 new pQTLs, demonstrating unique genetic regulation in CSF. We identified CSF-enriched pleiotropic regions on chromosome (chr)3q28 near OSTN and chr19q13.32 near APOE that were enriched for neuron specificity and neurological development. We integrated our associations with Alzheimer's disease (AD) through proteome-wide association study (PWAS), colocalization and Mendelian randomization and identified 38 putative causal proteins, 15 of which have drugs available. Finally, we developed a proteomics-based AD prediction model that outperforms genetics-based models. These findings will be instrumental to further understand the biology and identify causal and druggable proteins for brain and neurological traits.
Collapse
Affiliation(s)
- Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihua Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ciyang Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Chengran Yang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Bridget Phillips
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Yueyao Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Aleksandra Beric
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Priyanka Gorijala
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard J Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA
| | - Agustin Ruiz
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Marta Marquié
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Itziar de Rojas
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Jarod Rutledge
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Edward N Wilson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Betty Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Department of Psychiatry, Maastricht University, Maastricht, the Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, Amsterdam, the Netherlands
| | - Marta Del Campo Milan
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Ignacio Alvarez
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Miquel Aguilar
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
| | - Michael D Greicius
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Pau Pastor
- Memory Disorders Unit, Department of Neurology, University Hospital Mutua Terrassa, Terrassa, Spain
- Unit of Neurodegenerative Diseases, Department of Neurology, University Hospital Germans Trias i Pujol and the Germans Trias i Pujol Research Institute (IGTP) Badalona, Barcelona, Spain
| | | | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
19
|
Zheng JY, Pang RK, Ye JH, Su S, Shi J, Qiu YH, Pan HF, Zheng RY, Hu XR, Deng QW, Li XX, Cai YF, Zhang SJ. Huang-Lian-Jie-Du decoction alleviates cognitive impairment in high-fat diet-induced obese mice via Trem2/Dap12/Syk pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156248. [PMID: 39556986 DOI: 10.1016/j.phymed.2024.156248] [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: 04/23/2024] [Revised: 07/28/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Cognitive impairment induced by a high-fat diet (HFD) is common, but its mechanism is largely unknown. Huang-Lian-Jie-Du (HLJD) decoction is a classical and powerful prescription in China. It consists of four medicinal plants and is widely used in traditional Chinese medicines (TCM). Studies have shown that HLJD decoction is effective in treating obesity, depression, and so on. However, the therapeutic mechanism of HLJD is still poorly understood. PURPOSE Our study aimed to explore whether inflammatory factors and Trem2/Dap12/Syk pathway are involved in this process and whether HLJD treatment can repair cognitive impairment in HFD-induced obesity. METHODS To obtain the obese mice, male mice were treated with HFD (60 Kcal% fat) for 16 weeks. After an additional eight weeks, HLJD decoction was administered orally at doses of 4 and 8 g/kg daily for eight weeks. The mice were then subjected to four behavior tests. Aβ42, total Tau, inflammatory-related, and microglial dysregulation-related markers expression were measured. Molecular docking analysis was also conducted to predict the interaction of the chemical constituents of HLJD with human TREM2, DAP12, and SYK. HLJD at doses of 12.5, 25, and 50 µg/mL or limonin at concentrations of 12.5, 25, and 50 µM were used to treat BV2 cells for 24 h. CCK8 assay and Trem2, Dap12, Syk, and p-Syk expression were measured. RESULTS Our study revealed that cognitive impairment was evident in mice treated with HFD, indicating the impact of obesity on cognitive function. The expression of Aβ42 and total Tau in the hippocampus (HIP) was significantly higher in obese (HFD-V) mice compared to normal control (NC-V) mice. The Il6, Il1b, and Il10 mRNA expression levels were also markedly increased in the HIP of obese mice. Furthermore, Trem2, Dap12, p-Syk, and Iba1 expression were elevated in the HIP of obese mice. Importantly, HLJD treatment was found to repair cognitive impairment and lower the protein expression of Aβ42, Tau, Trem2, Dap12, p-Syk, and the expression of Il6, Il1b, and Il10 mRNA in HIP of HFD-V mice. The increased expression of Trem2, Dap12, p-Syk, and Iba1 in HIP after HFD consumption could be reduced after receiving HLJD decoction. The compound Limonin showed a well-predicted binding energy with TREM2, DAP12, and SYK. BV2 cells with HLJD or limonin detected the mRNA expressions of Trem2/Dap12. HLJD at 25 and 50 µg/mL decreased Trem2, Dap12, and p-Syk protein levels in BV2 cells. CONCLUSION These results reveal that HLJD treatment could alleviate cognitive impairment in HFD-induced obese mice by controlling the activation of the Trem2/Dap12 pathway and reducing Syk phosphorylation in HIP microglia. HLJD and limonin suppressed Trem2/Dap12/Syk signaling pathway in BV2 cells. HLJD therapy might represent a novel treatment for patients with cognitive impairment induced by obesity.
Collapse
Affiliation(s)
- Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Rui-Kang Pang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA
| | - Shan Su
- College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jia Shi
- College of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yu-Hui Qiu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210023, China
| | - Ru-Yu Zheng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Xin-Rui Hu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Qi-Wen Deng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Xiao-Xiao Li
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China.
| | - Ye-Feng Cai
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China.
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China; Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
20
|
Zhang Y, Guo X, Zhao J, Gao X, Zhang L, Huang T, Wang Y, Niu Q, Zhang Q. The downregulation of TREM2 exacerbates toxicity of development and neurobehavior induced by aluminum chloride and nano-alumina in adult zebrafish. Toxicol Appl Pharmacol 2024; 492:117107. [PMID: 39288838 DOI: 10.1016/j.taap.2024.117107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
To investigate the difference in the development and neurobehavior between aluminum chloride (AlCl3) and nano-alumina (AlNPs) in adult zebrafish and the role of triggering receptor expressed on myeloid cells (TREM2) in this process. Zebrafish embryos were randomly administered with control, negative control, TREM2 knockdown, AlCl3, TREM2 knockdown + AlCl3, AlNPs, and TREM2 knockdown + AlNPs, wherein AlCl3 and AlNPs were 50 mg/L and TREM2 knockdown was achieved by microinjecting lentiviral-containing TREM2 inhibitors into the yolk sac. We assessed development, neurobehavior, histopathology, ultrastructural structure, neurotransmitters (AChE, DA), SOD, genes of TREM2 and neurodevelopment (α1-tubulin, syn2a, mbp), and AD-related proteins and genes. AlCl3 significantly lowered the malformation rate than AlNPs, and further increased rates of malformation and mortality following TREM2 knockdown. The locomotor ability, learning and memory were similar between AlCl3 and AlNPs. TREM2 deficiency further exacerbated their impairment in panic reflex, microglia decrease, and nerve fibers thickening and tangling. AlCl3, rather than AlNPs, significantly elevated AChE activity and p-tau content while decreasing TREM2 and syn2a levels than the control. TREM2 loss further aggravated impairment in the AChE and SOD activity, and psen1 and p-tau levels. Therefore, AlCl3 induces greater developmental toxicity but equivalent neurobehavior toxicity than AlNPs, while their toxicity was intensified by TREM2 deficiency.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Toxicology, Shanxi Provincial Center for Disease Control and Prevention, Taiyuan 030012, China
| | - Xinyue Guo
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China; Department of Business Management, Shaanxi Provincial Center for Disease Control and Prevention, Xi'an 710054, China
| | - Jinjin Zhao
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaocheng Gao
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Lan Zhang
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Tao Huang
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Yanhong Wang
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Qiao Niu
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China
| | - Qinli Zhang
- Department of Occupational Medicine, School of public health, Shanxi Medical University, Taiyuan 030001, China; Department of Pathology, University of Mississippi Medical Center, 2500 N State St., Jackson, MS 39216, United States of America.
| |
Collapse
|
21
|
Zhang LL, Cheng P, Chu YQ, Zhou ZM, Hua R, Zhang YM. The microglial innate immune receptor TREM2 participates in fear memory formation through excessive prelimbic cortical synaptic pruning. Front Immunol 2024; 15:1412699. [PMID: 39544929 PMCID: PMC11560470 DOI: 10.3389/fimmu.2024.1412699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024] Open
Abstract
Introduction Fear memory formation has been implicated in fear- and stress-related psychiatric disorders, including post-traumatic stress disorder (PTSD) and phobias. Synapse deficiency and microglial activation are common among patients with PTSD, and induced in animal models of fear conditioning. Increasing studies now focus on explaining the specific mechanisms between microglia and synapse deficiency. Though newly-identified microglia regulator triggering receptor expressed on myeloid cells 2 (TREM2) plays a role in microglial phagocytic activity, its role in fear-formation remains unknown. Methods We successfully constructed a fear- formation model by foot-shock. Four days after foot-shock, microglial capacity of synaptic pruning was investigated via western blotting, immunofluorescence and Golgi-Cox staining. Prelimbic chemical deletion or microglia inhibition was performed to detect the role of microglia in synaptic loss and neuron activity. Finally, Trem2 knockout mice or wild-type mice with Trem2 siRNA injection were exposed to foot-shock to identify the involvement of TREM2 in fear memory formation. Results The results herein indicate that the foot-shock protocol in male mice resulted in a fear formation model. Mechanistically, fear conditioning enhanced the microglial capacity for engulfing synapse materials, and led to glutamatergic neuron activation in the prelimbic cortex. Prelimbic chemical deletion or microglia inhibition improved fear memory formation. Further investigation demonstrated that TREM2 regulates microglial phagocytosis, enhancing synaptic pruning. Trem2 knockout mice showed remarkable reductions in prelimbic synaptic pruning and reduced neuron activation, with decreased fear memory formation. Discussion Our cumulative results suggest that prelimbic TREM2-mediated excessive microglial synaptic pruning is involved in the fear memory formation process, leading to development of abnormal stress-related behavior.
Collapse
Affiliation(s)
- Le-le Zhang
- National Medical Products Administration Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Peng Cheng
- National Medical Products Administration Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Yuan-qing Chu
- National Medical Products Administration Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Zi-ming Zhou
- National Medical Products Administration Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Rong Hua
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong-mei Zhang
- National Medical Products Administration Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
22
|
Ming S, Li X, Xiao Q, Qu S, Wang Q, Fang Q, Liang P, Xu Y, Yang J, Yang Y, Huang X, Wu Y. TREM2 aggravates sepsis by inhibiting fatty acid oxidation via the SHP1/BTK axis. J Clin Invest 2024; 135:e159400. [PMID: 39405126 DOI: 10.1172/jci159400] [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: 02/15/2022] [Accepted: 10/08/2024] [Indexed: 01/03/2025] Open
Abstract
Impaired fatty acid oxidation (FAO) and the therapeutic benefits of FAO restoration have been revealed in sepsis. However, the regulatory factors contributing to FAO dysfunction during sepsis remain inadequately clarified. In this study, we identified a subset of lipid-associated macrophages characterized by high expression of trigger receptor expressed on myeloid cells 2 (TREM2) and demonstrated that TREM2 acted as a suppressor of FAO to increase the susceptibility to sepsis. TREM2 expression was markedly upregulated in sepsis patients and correlated with the severity of sepsis. Knockout of TREM2 in macrophages improved the survival rate and reduced inflammation and organ injuries of sepsis mice. Notably, TREM2-deficient mice exhibited decreased triglyceride accumulation and an enhanced FAO rate. Further observations showed that the blockade of FAO substantially abolished the alleviated symptoms observed in TREM2-knockout mice. Mechanically, we demonstrated that TREM2 interacted with the phosphatase SHP1 to inhibit bruton tyrosine kinase-mediated (BTK-mediated) FAO in sepsis. Our findings expand the understanding of FAO dysfunction in sepsis and reveal TREM2 as a critical regulator of FAO that may provide a promising target for the clinical treatment of sepsis.
Collapse
Affiliation(s)
- Siqi Ming
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
| | - Qiang Xiao
- Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Siying Qu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qiaohua Wang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Qiongyan Fang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Pingping Liang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Yating Xu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People' s Hospital, the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jingwen Yang
- Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Yongqiang Yang
- Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai Hospital, Zhuhai, China
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People' s Hospital, the Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Key Research Laboratory of Traditional Chinese Medicine in the Prevention and Treatment of Infectious Diseases, Traditional Chinese Medicine Bureau of Guangdong Province, the Fifth Affiliated Hospital, SunYat-Sen University, Zhuhai, China
| |
Collapse
|
23
|
James R, Subramanyam KN, Payva F, E AP, Tv VK, Sivaramakrishnan V, Ks S. In-silico analysis predicts disruption of normal angiogenesis as a causative factor in osteoporosis pathogenesis. BMC Genom Data 2024; 25:85. [PMID: 39379846 PMCID: PMC11460074 DOI: 10.1186/s12863-024-01269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
Angiogenesis-osteogenesis coupling is critical for proper functioning and maintaining the health of bones. Any disruption in this coupling, associated with aging and disease, might lead to loss of bone mass. Osteoporosis (OP) is a debilitating bone metabolic disorder that affects the microarchitecture of bones, gradually leading to fracture. Computational analysis revealed that normal angiogenesis is disrupted during the progression of OP, especially postmenopausal osteoporosis (PMOP). The genes associated with OP and PMOP were retrieved from the DisGeNET database. Hub gene analysis and molecular pathway enrichment were performed via the Cytoscape plugins STRING, MCODE, CytoHubba, ClueGO and the web-based tool Enrichr. Twenty-eight (28) hub genes were identified, eight of which were transcription factors (HIF1A, JUN, TP53, ESR1, MYC, PPARG, RUNX2 and SOX9). Analysis of SNPs associated with hub genes via the gnomAD, I-Mutant2.0, MUpro, ConSurf and COACH servers revealed the substitution F201L in IL6 as the most deleterious. The IL6 protein was modeled in the SWISS-MODEL server and the substitution was analyzed via the YASARA FoldX plugin. A positive ΔΔG (1.936) of the F201L mutant indicates that the mutated structure is less stable than the wild-type structure is. Thirteen hub genes, including IL6 and the enriched molecular pathways were found to be profoundly involved in angiogenesis/endothelial function and immune signaling. Mechanical loading of bones through weight-bearing exercises can activate osteoblasts via mechanotransduction leading to increased bone formation. The present study suggests proper mechanical loading of bone as a preventive strategy for PMOP, by which angiogenesis and the immune status of the bone can be maintained. This in silico analysis could be used to understand the molecular etiology of OP and to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Remya James
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India.
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| | - Koushik Narayan Subramanyam
- Department of Orthopaedics, Sri Sathya Sai Institute of Higher Medical Sciences, Prasanthigram, Puttaparthi, Andhra Pradesh, 515134, India
| | - Febby Payva
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India
| | - Amrisa Pavithra E
- Department of Zoology, St. Joseph's College for Women, Alappuzha, Kerala, 688001, India
| | - Vineeth Kumar Tv
- Department of Zoology, The Cochin College, Kochi, Kerala, 682002, India.
| | - Venketesh Sivaramakrishnan
- School of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthinilayam, Puttaparthi, Andhra Pradesh, 515134, India
| | - Santhy Ks
- School of Biosciences, Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore, Tamil Nadu, 614043, India.
| |
Collapse
|
24
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
25
|
Früholz I, Meyer-Luehmann M. The intricate interplay between microglia and adult neurogenesis in Alzheimer's disease. Front Cell Neurosci 2024; 18:1456253. [PMID: 39360265 PMCID: PMC11445663 DOI: 10.3389/fncel.2024.1456253] [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: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system, play a crucial role in regulating adult neurogenesis and contribute significantly to the pathogenesis of Alzheimer's disease (AD). Under physiological conditions, microglia support and modulate neurogenesis through the secretion of neurotrophic factors, phagocytosis of apoptotic cells, and synaptic pruning, thereby promoting the proliferation, differentiation, and survival of neural progenitor cells (NPCs). However, in AD, microglial function becomes dysregulated, leading to chronic neuroinflammation and impaired neurogenesis. This review explores the intricate interplay between microglia and adult neurogenesis in health and AD, synthesizing recent findings to provide a comprehensive overview of the current understanding of microglia-mediated regulation of adult neurogenesis. Furthermore, it highlights the potential of microglia-targeted therapies to modulate neurogenesis and offers insights into potential avenues for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Iris Früholz
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
26
|
Liang J, Menon A, Tomco T, Bhattarai N, Smith IN, Khrestian M, Formica SV, Eng C, Buck M, Bekris LM. A Computational Approach in the Systematic Search of the Interaction Partners of Alternatively Spliced TREM2 Isoforms. Int J Mol Sci 2024; 25:9667. [PMID: 39273614 PMCID: PMC11395018 DOI: 10.3390/ijms25179667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease is the most common form of dementia, characterized by the pathological accumulation of amyloid-beta (Aβ) plaques and tau neurofibrillary tangles. Triggering receptor expressed on myeloid cells 2 (TREM2) is increasingly recognized as playing a central role in Aβ clearance and microglia activation in AD. The TREM2 gene transcriptional product is alternatively spliced to produce three different protein isoforms. The canonical TREM2 isoform binds to DAP12 to activate downstream pathways. However, little is known about the function or interaction partners of the alternative TREM2 isoforms. The present study utilized a computational approach in a systematic search for new interaction partners of the TREM2 isoforms by integrating several state-of-the-art structural bioinformatics tools from initial large-scale screening to one-on-one corroborative modeling and eventual all-atom visualization. CD9, a cell surface glycoprotein involved in cell-cell adhesion and migration, was identified as a new interaction partner for two TREM2 isoforms, and CALM, a calcium-binding protein involved in calcium signaling, was identified as an interaction partner for a third TREM2 isoform, highlighting the potential role of cell adhesion and calcium regulation in AD.
Collapse
Affiliation(s)
- Junyi Liang
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
| | - Aditya Menon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Taylor Tomco
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
| | - Nisha Bhattarai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Iris Nira Smith
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
| | - Maria Khrestian
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
| | - Shane V. Formica
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (J.L.); (T.T.); (I.N.S.); (M.K.); (S.V.F.)
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
27
|
Matteoli M. The role of microglial TREM2 in development: A path toward neurodegeneration? Glia 2024; 72:1544-1554. [PMID: 38837837 DOI: 10.1002/glia.24574] [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: 01/20/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
The nervous and the immune systems undergo a continuous cross talk, starting from early development and continuing throughout adulthood and aging. Defects in this cross talk contribute to neurodevelopmental and neurodegenerative diseases. Microglia are the resident immune cells in the brain that are primarily involved in this bidirectional communication. Among the microglial genes, trem2 is a key player, controlling the functional state of microglia and being at the forefront of many processes that require interaction between microglia and other brain components, such as neurons and oligodendrocytes. The present review focuses on the early developmental window, describing the early brain processes in which TREM2 is primarily involved, including the modulation of synapse formation and elimination, the control of neuronal bioenergetic states as well as the contribution to myelination processes and neuronal circuit formation. By causing imbalances during these early maturation phases, dysfunctional TREM2 may have a striking impact on the adult brain, making it a more sensitive target for insults occurring during adulthood and aging.
Collapse
Affiliation(s)
- Michela Matteoli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
28
|
Zhang M, Duan Y, Gan H, Jiang N, Qin L, Luo Y, Palahati A, He Y, Li C, Zhai X. TYROBP serve as potential immune-related signature genes in the acute phase of intracerebral hemorrhage. Sci Rep 2024; 14:20158. [PMID: 39215129 PMCID: PMC11364555 DOI: 10.1038/s41598-024-71132-9] [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: 01/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The development of intracerebral hemorrhage (ICH) is a dynamic process and intervention during the acute phase of ICH is critical for subsequent recovery. Therefore, it is crucial to screen potential signature genes and therapeutic target genes in the acute phase of ICH. In this study, based on the results of mRNA sequencing in mouse ICH and mRNA sequencing of human ICH from online databases, top five potential signature genes after ICH, Tyrobp, Itgb2, Tlr2, Ptprc and Itgam, were screened. Quantitative PCR results showed higher mRNA expression of Tyrobp, Itgb2, Tlr2, Ptprc, and Itgam in the 1-, 3- and 5-day mouse ICH groups compared to the sham-operated group. Immune infiltration correlation analysis shows that the top-ranked signature gene, Tyrobp, is negatively correlated with M2 macrophages and plasma cells, and Western blot analysis shows higher expression of the Tyrobp protein in the 1-, 3-, and 5-day mouse ICH groups compared to the sham-operated group. Furthermore, immunohistochemistry revealed that TYROBP protein expression was significantly higher in human ICH tissues than in normal brain tissues. Our results suggest that Tyrobp is a signature gene in the acute phase of ICH and may be a potential target for the treatment of the acute phase of ICH.
Collapse
Affiliation(s)
- Mi Zhang
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yuhao Duan
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hui Gan
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing, 400016, China
| | - Le Qin
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yujia Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ailiyaer Palahati
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yaying He
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Chenyang Li
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xuan Zhai
- Department of Neurosurgery, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 401122, China.
| |
Collapse
|
29
|
Hwang M, Bergmann CC. Neurotropic murine coronavirus mediated demyelination: Factors dampening pathogenesis. J Neuroimmunol 2024; 393:578382. [PMID: 38850674 DOI: 10.1016/j.jneuroim.2024.578382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Virus infections and autoimmune responses are implicated as primary triggers of demyelinating diseases. Specifically, the association of Epstein-Barr virus (EBV) infection with development of multiple sclerosis (MS) has re-ignited an interest in virus induced autoimmune responses to CNS antigens. Nevertheless, demyelination may also be caused by immune mediated bystander pathology in an attempt to control direct infection in the CNS. Tissue damage as a result of anti-viral responses or low level viral persistence may lead to immune activation manifesting in demyelinating lesions, axonal damage and clinical symptoms. This review focuses on the neurotropic mouse coronavirus induced demyelination model to highlight how immune responses activated during the acute phase pave the way to dampen pathology and promote repair. We specifically discuss the role of immune dampening factors programmed cell death ligand 1 (PD-L1) and interleukin (IL)-10, as well as microglia and triggering receptor expressed on myeloid cells 2 (Trem2), in limiting demyelination independent of viral persistence.
Collapse
Affiliation(s)
- Mihyun Hwang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
30
|
O'Shea TM, Ao Y, Wang S, Ren Y, Cheng AL, Kawaguchi R, Shi Z, Swarup V, Sofroniew MV. Derivation and transcriptional reprogramming of border-forming wound repair astrocytes after spinal cord injury or stroke in mice. Nat Neurosci 2024; 27:1505-1521. [PMID: 38907165 PMCID: PMC11303254 DOI: 10.1038/s41593-024-01684-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 05/15/2024] [Indexed: 06/23/2024]
Abstract
Central nervous system (CNS) lesions become surrounded by neuroprotective borders of newly proliferated reactive astrocytes; however, fundamental features of these cells are poorly understood. Here we show that following spinal cord injury or stroke, 90% and 10% of border-forming astrocytes derive, respectively, from proliferating local astrocytes and oligodendrocyte progenitor cells in adult mice of both sexes. Temporal transcriptome analysis, single-nucleus RNA sequencing and immunohistochemistry show that after focal CNS injury, local mature astrocytes dedifferentiate, proliferate and become transcriptionally reprogrammed to permanently altered new states, with persisting downregulation of molecules associated with astrocyte-neuron interactions and upregulation of molecules associated with wound healing, microbial defense and interactions with stromal and immune cells. These wound repair astrocytes share morphologic and transcriptional features with perimeningeal limitans astrocytes and are the predominant source of neuroprotective borders that re-establish CNS integrity around lesions by separating neural parenchyma from stromal and immune cells as occurs throughout the healthy CNS.
Collapse
Affiliation(s)
- Timothy M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shinong Wang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yilong Ren
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Amy L Cheng
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, CA, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Li H, Guan M, Zhang NN, Wang Y, Liang T, Wu H, Wang C, Sun T, Liu S. Harnessing nanomedicine for modulating microglial states in the central nervous system disorders: Challenges and opportunities. Biomed Pharmacother 2024; 177:117011. [PMID: 38917758 DOI: 10.1016/j.biopha.2024.117011] [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: 03/12/2024] [Revised: 05/30/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia are essential for maintaining homeostasis and responding to pathological events in the central nervous system (CNS). Their dynamic and multidimensional states in different environments are pivotal factors in various CNS disorders. However, therapeutic modulation of microglial states is challenging due to the intricate balance these cells maintain in the CNS environment and the blood-brain barrier's restriction of drug delivery. Nanomedicine presents a promising avenue for addressing these challenges, offering a method for the targeted and efficient modulation of microglial states. This review covers the challenges faced in microglial therapeutic modulation and potential use of nanoparticle-based drug delivery systems. We provide an in-depth examination of nanoparticle applications for modulating microglial states in a range of CNS disorders, encompassing neurodegenerative and autoimmune diseases, infections, traumatic injuries, stroke, tumors, chronic pain, and psychiatric conditions. This review highlights the recent advancements and future prospects in nanomedicine for microglial modulation, paving the way for future research and clinical applications of therapeutic interventions in CNS disorders.
Collapse
Affiliation(s)
- Haisong Li
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Department of Neurosurgery, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Meng Guan
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China
| | - Yizhuo Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Tingting Liang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Haitao Wu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Chang Wang
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China; International Center of Future Science, Jilin University, Changchun, Jilin, China; State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin, China.
| | - Shuhan Liu
- Cancer Center, The First Hospital, Jilin University, Changchun, Jilin, China; Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, Jilin, China; National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin, China.
| |
Collapse
|
32
|
Mi X, Kang C, Hou S, Gao Y, Hao L, Gao X. Mining and exploration of appendicitis nursing targets: An observational study. Medicine (Baltimore) 2024; 103:e38667. [PMID: 38941398 PMCID: PMC11466127 DOI: 10.1097/md.0000000000038667] [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: 05/10/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024] Open
Abstract
Appendicitis is an inflammation caused by obstruction of the appendiceal lumen or termination of blood supply leading to appendiceal necrosis followed by secondary bacterial infection. The relationship between TYROBP gene and the nursing of appendicitis remains unclear. The appendicitis dataset GSE9579 profile was downloaded from the gene expression omnibus database generated from GPL571. Differentially expressed genes were screened, followed by weighted gene co-expression network analysis, functional enrichment analysis, gene set enrichment analysis, construction and analysis of protein-protein interaction network, Comparative Toxicogenomics Database analysis, and immune infiltration analysis. Heatmaps of gene expression levels were plotted. A total of 1570 differentially expressed genes were identified. According to gene ontology analysis, they were mainly enriched in organic acid metabolic process, condensed chromosome kinetochore, oxidoreductase activity. In Kyoto Encyclopedia of Gene and Genome analysis, they mainly concentrated in metabolic pathways, P53 signaling pathway, PPAR signaling pathway. The soft threshold power in weighted gene co-expression network analysis was set to 12. Through the construction and analysis of protein-protein interaction network, 5 core genes (FCGR2A, IL1B, ITGAM, TLR2, TYROBP) were obtained. Heatmap of core gene expression levels revealed high expression of TYROBP in appendicitis samples. Comparative Toxicogenomics Database analysis found that core genes (FCGR2A, IL1B, ITGAM, TLR2, TYROBP) were closely related to abdominal pain, gastrointestinal dysfunction, fever, and inflammation occurrence. TYROBP gene is highly expressed in appendicitis, and higher expression of TYROBP gene indicates worse prognosis. TYROBP may serve as a molecular target for appendicitis and its nursing.
Collapse
Affiliation(s)
- Xihua Mi
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Chunbo Kang
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Shiyang Hou
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Yanfang Gao
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Lingli Hao
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Gao
- Gastrointestinal Rehabilitation Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Bharadwaj S, Groza Y, Mierzwicka JM, Malý P. Current understanding on TREM-2 molecular biology and physiopathological functions. Int Immunopharmacol 2024; 134:112042. [PMID: 38703564 DOI: 10.1016/j.intimp.2024.112042] [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: 02/08/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 05/06/2024]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM-2), a glycosylated receptor belonging to the immunoglobin superfamily and especially expressed in the myeloid cell lineage, is frequently explained as a reminiscent receptor for both adaptive and innate immunity regulation. TREM-2 is also acknowledged to influence NK cell differentiation via the PI3K and PLCγ signaling pathways, as well as the partial activation or direct inhibition of T cells. Additionally, TREM-2 overexpression is substantially linked to cell-specific functions, such as enhanced phagocytosis, reduced toll-like receptor (TLR)-mediated inflammatory cytokine production, increased transcription of anti-inflammatory cytokines, and reshaped T cell function. Whereas TREM-2-deficient cells exhibit diminished phagocytic function and enhanced proinflammatory cytokines production, proceeding to inflammatory injuries and an immunosuppressive environment for disease progression. Despite the growing literature supporting TREM-2+ cells in various diseases, such as neurodegenerative disorders and cancer, substantial facets of TREM-2-mediated signaling remain inadequately understood relevant to pathophysiology conditions. In this direction, herein, we have summarized the current knowledge on TREM-2 biology and cell-specific TREM-2 expression, particularly in the modulation of pivotal TREM-2-dependent functions under physiopathological conditions. Furthermore, molecular regulation and generic biological relevance of TREM-2 are also discussed, which might provide an alternative approach for preventing or reducing TREM-2-associated deformities. At last, we discussed the TREM-2 function in supporting an immunosuppressive cancer environment and as a potential drug target for cancer immunotherapy. Hence, summarized knowledge of TREM-2 might provide a window to overcome challenges in clinically effective therapies for TREM-2-induced diseases in humans.
Collapse
Affiliation(s)
- Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| | - Yaroslava Groza
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Joanna M Mierzwicka
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Center, Průmyslová 595, 252 50 Vestec, Czech Republic.
| |
Collapse
|
34
|
Yan H, Wang W, Cui T, Shao Y, Li M, Fang L, Feng L. Advances in the Understanding of the Correlation Between Neuroinflammation and Microglia in Alzheimer's Disease. Immunotargets Ther 2024; 13:287-304. [PMID: 38881647 PMCID: PMC11180466 DOI: 10.2147/itt.s455881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease with a subtle and progressive onset and is the most common type of dementia. However, its etiology and pathogenesis have not yet been fully elucidated. The common pathological manifestations of AD include extraneuronal β-amyloid deposition (Aβ), intraneuronal tau protein phosphorylation leading to the formation of 'neurofibrillary tangles' (NFTs), neuroinflammation, progressive loss of brain neurons/synapses, and glucose metabolism disorders. Current treatment approaches for AD primarily focus on the 'Aβ cascade hypothesis and abnormal aggregation of hyperphosphorylation of tau proteins', but have shown limited efficacy. Therefore, there is an ongoing need to identify more effective treatment targets for AD. The central nervous system (CNS) inflammatory response plays a key role in the occurrence and development of AD. Neuroinflammation is an immune response activated by glial cells in the CNS that usually occurs in response to stimuli such as nerve injury, infection and toxins or in response to autoimmunity. Neuroinflammation ranks as the third most prominent pathological feature in AD, following Aβ and NFTs. In recent years, the focus on the role of neuroinflammation and microglia in AD has increased due to the advancements in genome-wide association studies (GWAS) and sequencing technology. Furthermore, research has validated the pivotal role of microglia-mediated neuroinflammation in the progression of AD. Therefore, this article reviews the latest research progress on the role of neuroinflammation triggered by microglia in AD in recent years, aiming to provide a new theoretical basis for further exploring the role of neuroinflammation in the process of AD occurrence and development.
Collapse
Affiliation(s)
- Huiying Yan
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Wei Wang
- Department of Intensive Care Unit, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Tingting Cui
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Yanxin Shao
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Limei Fang
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| |
Collapse
|
35
|
Yang R, Li DD, Li XX, Yang XX, Gao HM, Zhang F. Dihydroquercetin alleviates dopamine neuron loss via regulating TREM2 activation. Int J Biol Macromol 2024; 269:132179. [PMID: 38723817 DOI: 10.1016/j.ijbiomac.2024.132179] [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: 03/16/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a prevalent neurodegenerative disorder, marked by the degeneration of dopamine (DA) neurons in the substantia nigra (SN). Current evidence strongly suggests that neuroinflammation, primarily mediated by microglia, contributes to PD pathogenesis. Triggering receptor expressed on myeloid cells 2 (TREM2) might serve as a promising therapeutic target for PD due to its ability to suppress neuroinflammation. Dihydroquercetin (DHQ) is an important natural dihydroflavone and confers apparent anti-inflammatory, antioxidant and anti-fibrotic effects. Recently, DHQ-mediated neuroprotection was exhibited. However, the specific mechanisms of its neuroprotective effects remain incompletely elucidated. METHODS In this study, rat models were utilized to induce damage to DA neurons using lipopolysaccharide (LPS) and 6-hydroxydopamine (6-OHDA) to assess the impacts of DHQ on the loss of DA neurons. Furthermore, DA neuronal MN9D cells and microglial BV2 cells were employed to investigate the function of TREM2 in DHQ-mediated DA neuroprotection. Finally, TREM2 knockout mice were used to investigate whether the neuroprotective effects mediated by DHQ through a mechanism dependent on TREM2. RESULTS The main findings demonstrated that DHQ effectively protected DA neurons against neurotoxicity induced by LPS and 6-OHDA and inhibited microglia-elicited neuroinflammation. Meanwhile, DHQ promoted microglial TREM2 signaling activation. Notably, DHQ failed to reduce inflammatory cytokines release and further present neuroprotection from DA neurotoxicity upon TREM2 silencing. Similarly, DHQ didn't exert DA neuroprotection in TREM2 knockout mice. CONCLUSIONS These findings suggest that DHQ exerted DA neuroprotection by regulating microglia TREM2 activation.
Collapse
Affiliation(s)
- Rong Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Dai-di Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao-Xian Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin-Xing Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Hui-Ming Gao
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Institute for Brain Sciences, Jiangsu Key Laboratory of Molecular Medicine, Model Animal Research Center, School of medicine, Nanjing University, Nanjing, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
36
|
Zhou X, Song H, He J, Han W, Li Q. Deciphering microglial activation and neuronal apoptosis post‑traumatic brain injury: The role of TYROBP in inflammation regulation networks. Mol Med Rep 2024; 29:104. [PMID: 38639190 PMCID: PMC11063751 DOI: 10.3892/mmr.2024.13228] [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: 11/30/2023] [Accepted: 02/01/2024] [Indexed: 04/20/2024] Open
Abstract
Traumatic Brain Injury (TBI) represents a significant public health challenge. Recovery from brain injury necessitates the collaborative efforts of various resident neural cells, predominantly microglia. The present study analyzed rat and mouse RNA expression micro‑arrays, high‑throughput RNA sequencing and single‑cell sequencing data sourced from public databases. To construct an inflammation regulation network around TYRO protein tyrosine kinase‑binding protein (TYROBP), to evaluate the role of TYROBP in cell death after TBI. These findings indicate that following TBI, neurons predominantly communicate with one another through the CXC chemokine ligand (CXCL) and CC chemokine ligand (CCL) signaling pathways, employing a paracrine mechanism to activate microglia. These activated microglia intensify the pathological progression of brain injury by releasing factors such as tumor necrosis factor α (TNF‑α), vascular endothelial growth factor and transforming growth factor β via the NF‑κB pathway. Cells co‑culture experiments demonstrated that neurons, impaired by mechanical injury, interact with microglia through non‑contact mechanisms. Activated microglia secrete cytokines, including TNF‑α, CXCL‑8 and CCL2, which trigger an inflammatory response and facilitate neuronal apoptosis. TYROBP gene knockout in microglia was demonstrated to reduce this interaction and reduce neuronal cell apoptosis rates.
Collapse
Affiliation(s)
- Xudong Zhou
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Huiping Song
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Jingjing He
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Wei Han
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Qin Li
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
37
|
Barry-Carroll L, Gomez-Nicola D. The molecular determinants of microglial developmental dynamics. Nat Rev Neurosci 2024; 25:414-427. [PMID: 38658739 DOI: 10.1038/s41583-024-00813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
Microglia constitute the largest population of parenchymal macrophages in the brain and are considered a unique subset of central nervous system glial cells owing to their extra-embryonic origins in the yolk sac. During development, microglial progenitors readily proliferate and eventually colonize the entire brain. In this Review, we highlight the origins of microglial progenitors and their entry routes into the brain and discuss the various molecular and non-molecular determinants of their fate, which may inform their specific functions. Specifically, we explore recently identified mechanisms that regulate microglial colonization of the brain, including the availability of space, and describe how the expansion of highly proliferative microglial progenitors facilitates the occupation of the microglial niche. Finally, we shed light on the factors involved in establishing microglial identity in the brain.
Collapse
Affiliation(s)
- Liam Barry-Carroll
- Nutrineuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Bordeaux, France
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
38
|
Fremuth LE, Hu H, van de Vlekkert D, Annunziata I, Weesner JA, Gomero E, d'Azzo A. Neuraminidase 1 regulates the cellular state of microglia by modulating the sialylation of Trem2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595036. [PMID: 38826426 PMCID: PMC11142087 DOI: 10.1101/2024.05.20.595036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Neuraminidase 1 (Neu1) cleaves terminal sialic acids from sialoglycoproteins in endolysosomes and at the plasma membrane. As such, Neu1 regulates immune cells, primarily those of the monocytic lineage. Here we examined how Neu1 influences microglia by modulating the sialylation of full-length Trem2 (Trem2-FL), a multifunctional receptor that regulates microglial survival, phagocytosis, and cytokine production. When Neu1 was deficient/downregulated, Trem2-FL remained sialylated, accumulated intracellularly, and was excessively cleaved into a C-terminal fragment (Trem2-CTF) and an extracellular soluble domain (sTrem2), enhancing their signaling capacities. Sialylated Trem2-FL (Sia-Trem2-FL) did not hinder Trem2-FL-DAP12-Syk complex assembly but impaired signal transduction through Syk, ultimately abolishing Trem2-dependent phagocytosis. Concurrently, Trem2-CTF-DAP12 complexes dampened NFκB signaling, while sTrem2 propagated Akt-dependent cell survival and NFAT1-mediated production of TNFα and CCL3. Because Neu1 and Trem2 are implicated in neurodegenerative/neuroinflammatory diseases, including Alzheimer disease (AD) and sialidosis, modulating Neu1 activity represents a therapeutic approach to broadly regulate microglia-mediated neuroinflammation.
Collapse
|
39
|
Serrano PDL, Rodrigues TDPV, Pinto LD, Pereira IC, Farias IB, Cavalheiro RBR, Mendes PM, Peixoto KO, Barile JP, Seneor DD, Correa Silva EG, Oliveira ASB, Pinto WBVDR, Sgobbi P. Assessing Chitinases and Neurofilament Light Chain as Biomarkers for Adult-Onset Leukodystrophies. Curr Issues Mol Biol 2024; 46:4309-4323. [PMID: 38785530 PMCID: PMC11120026 DOI: 10.3390/cimb46050262] [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: 03/31/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Leukodystrophies represent a large and complex group of inherited disorders affecting the white matter of the central nervous system. Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) is a rare leukodystrophy which still needs the proper identification of diagnostic, prognostic, and monitoring biomarkers. The aim of this study was to determine the diagnostic and prognostic value of chitinases and neurofilament light chain as biomarkers for ALSP. A cross-sectional study was performed to analyze cerebrospinal fluid levels of chitinases (chitotriosidase and chitinase 3-like 2) and neurofilament light chain in five different groups: (i) normal health individuals; (ii) patients with definitive diagnosis of ALSP and genetic confirmation; (iii) asymptomatic patients with CSF1R variants; (iv) patients with other adult-onset leukodystrophies; and (v) patients with amyotrophic lateral sclerosis (external control group). Chitinase levels showed a statistical correlation with clinical assessment parameters in ALSP patients. Chitinase levels were also distinct between ALSP and the other leukodystrophies. Significant differences were noted in the levels of chitinases and neurofilament light chain comparing symptomatic (ALSP) and asymptomatic individuals with CSF1R variants. This study is the first to establish chitinases as a potential biomarker for ALSP and confirms neurofilament light chain as a good biomarker for primary microgliopathies.
Collapse
Affiliation(s)
- Paulo de Lima Serrano
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | | | - Leslyê Donato Pinto
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
| | - Indiara Correia Pereira
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
| | - Igor Braga Farias
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Renan Brandão Rambaldi Cavalheiro
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Patrícia Marques Mendes
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Kaliny Oliveira Peixoto
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - João Paulo Barile
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Daniel Delgado Seneor
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | | | - Acary Souza Bulle Oliveira
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Wladimir Bocca Vieira de Rezende Pinto
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| | - Paulo Sgobbi
- PSEG Centro de Pesquisa Clínica, São Paulo 04038-002, SP, Brazil; (P.d.L.S.); (T.d.P.V.R.); (L.D.P.); (I.C.P.); (E.G.C.S.)
- Department of Neurology and Neurosurgery, Federal University of São Paulo (UNIFESP), São Paulo 04039-060, SP, Brazil; (I.B.F.); (R.B.R.C.); (P.M.M.); (K.O.P.); (J.P.B.); (D.D.S.); (A.S.B.O.); (W.B.V.d.R.P.)
| |
Collapse
|
40
|
Homma H, Yoshioka Y, Fujita K, Shirai S, Hama Y, Komano H, Saito Y, Yabe I, Okano H, Sasaki H, Tanaka H, Okazawa H. Dynamic molecular network analysis of iPSC-Purkinje cells differentiation delineates roles of ISG15 in SCA1 at the earliest stage. Commun Biol 2024; 7:413. [PMID: 38594382 PMCID: PMC11003991 DOI: 10.1038/s42003-024-06066-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 03/18/2024] [Indexed: 04/11/2024] Open
Abstract
Better understanding of the earliest molecular pathologies of all neurodegenerative diseases is expected to improve human therapeutics. We investigated the earliest molecular pathology of spinocerebellar ataxia type 1 (SCA1), a rare familial neurodegenerative disease that primarily induces death and dysfunction of cerebellum Purkinje cells. Extensive prior studies have identified involvement of transcription or RNA-splicing factors in the molecular pathology of SCA1. However, the regulatory network of SCA1 pathology, especially central regulators of the earliest developmental stages and inflammatory events, remains incompletely understood. Here, we elucidated the earliest developmental pathology of SCA1 using originally developed dynamic molecular network analyses of sequentially acquired RNA-seq data during differentiation of SCA1 patient-derived induced pluripotent stem cells (iPSCs) to Purkinje cells. Dynamic molecular network analysis implicated histone genes and cytokine-relevant immune response genes at the earliest stages of development, and revealed relevance of ISG15 to the following degradation and accumulation of mutant ataxin-1 in Purkinje cells of SCA1 model mice and human patients.
Collapse
Affiliation(s)
- Hidenori Homma
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yuki Yoshioka
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kyota Fujita
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Research Center for Child Mental Development, Kanazawa University, 13-1 Takaramachi, Kanazawa-shi, Ishikawa, 920-8640, Japan
| | - Shinichi Shirai
- Department of Neurology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yuka Hama
- Department of Neurology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hajime Komano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuko Saito
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidenao Sasaki
- Department of Neurology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hikari Tanaka
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
41
|
Magni G, Riboldi B, Ceruti S. Human Glial Cells as Innovative Targets for the Therapy of Central Nervous System Pathologies. Cells 2024; 13:606. [PMID: 38607045 PMCID: PMC11011741 DOI: 10.3390/cells13070606] [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: 03/11/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
In vitro and preclinical in vivo research in the last 35 years has clearly highlighted the crucial physiopathological role of glial cells, namely astrocytes/microglia/oligodendrocytes and satellite glial cells/Schwann cells in the central and peripheral nervous system, respectively. Several possible pharmacological targets to various neurodegenerative disorders and painful conditions have therefore been successfully identified, including receptors and enzymes, and mediators of neuroinflammation. However, the translation of these promising data to a clinical setting is often hampered by both technical and biological difficulties, making it necessary to perform experiments on human cells and models of the various diseases. In this review we will, therefore, summarize the most relevant data on the contribution of glial cells to human pathologies and on their possible pharmacological modulation based on data obtained in post-mortem tissues and in iPSC-derived human brain cells and organoids. The possibility of an in vivo visualization of glia reaction to neuroinflammation in patients will be also discussed.
Collapse
Affiliation(s)
| | | | - Stefania Ceruti
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milan, Italy; (G.M.); (B.R.)
| |
Collapse
|
42
|
Wang X, Wang J, Chen Y, Qian X, Luo S, Wang X, Ma C, Ge W. The aldehyde dehydrogenase 2 rs671 variant enhances amyloid β pathology. Nat Commun 2024; 15:2594. [PMID: 38519490 PMCID: PMC10959958 DOI: 10.1038/s41467-024-46899-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
In the ALDH2 rs671 variant, a guanine changes to an adenine, resulting in a dramatic decrease in the catalytic activity of the enzyme. Population-based data are contradictory about whether this variant increases the risk of Alzheimer's disease. In East Asian populations, the prevalence of the ALDH2 rs671 variant is 30-50%, making the National Human Brain Bank for Development and Function (the largest brain bank in East Asia) an important resource to explore the link between the ALDH2 rs671 polymorphism and Alzheimer's disease pathology. Here, using 469 postmortem brains, we find that while the ALDH2 rs671 variant is associated with increased plaque deposits and a higher Aβ40/42 ratio, it is not an independent risk factor for Alzheimer's disease. Mechanistically, we show that lower ALDH2 activity leads to 4-HNE accumulation in the brain. The (R)-4-HNE enantiomer adducts to residue Lys53 of C99, favoring Aβ40 generation in the Golgi apparatus. Decreased ALDH2 activity also lowers inflammatory factor secretion, as well as amyloid β phagocytosis and spread in brains of patients with Alzheimer's disease. We thus define the relationship between the ALDH2 rs671 polymorphism and amyloid β pathology, and find that ALDH2 rs671 is a key regulator of Aβ40 or Aβ42 generation.
Collapse
Affiliation(s)
- Xia Wang
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiayu Wang
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yashuang Chen
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiaojing Qian
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Shiqi Luo
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xue Wang
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Wei Ge
- Department of Immunology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| |
Collapse
|
43
|
Zheng P, Tan Y, Liu Q, Wu C, Kang J, Liang S, Zhu L, Yan K, Zeng L, Chen B. Deciphering the molecular and clinical characteristics of TREM2, HCST, and TYROBP in cancer immunity: A comprehensive pan-cancer study. Heliyon 2024; 10:e26993. [PMID: 38468942 PMCID: PMC10926084 DOI: 10.1016/j.heliyon.2024.e26993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/23/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Background Hematopoietic cell signal transducer (HCST) and tyrosine kinase-binding protein (TYROBP) are triggering receptors expressed on myeloid cells 2 (TREM2), which are pivotal in the immune response to disease. Despite growing evidence underscoring the significance of TREM2, HCST, and TYROBP in certain forms of tumorigenesis, a comprehensive pan-cancer analysis of these proteins is lacking. Methods Multiple databases were synthesized to investigate the relationship between TREM2, HCST, TYROBP, and various cancer types. These include prognosis, methylation, regulation by long non-coding RNAs and transcription factors, immune signatures, pathway activity, microsatellite instability (MSI), tumor mutational burden (TMB), single-cell transcriptome profiling, and drug sensitivity. Results TREM2, HCST, and TYROBP displayed extensive somatic changes across numerous tumors, and their mRNA expression and methylation levels influenced patient outcomes across multiple cancer types. long non-coding RNA (lncRNA) -messenger RNA (mRNA) and TF-mRNA regulatory networks involving TREM2, HCST, and TYROBP were identified, with lncRNA MEG3 and the transcription factor SIP1 emerging as potential key regulators. Further immune analyses indicated that TREM2, HCST, and TYROBP play critical roles in immune-related pathways and macrophage differentiation, and may be significantly associated with TGF-β and SMAD9. Furthermore, the expression of TREM2, HCST, and TYROBP correlated with the immunotherapy markers TMB and MSI, and influenced sensitivity to immune-targeted drugs, thereby indicating their potential as predictors of immunotherapy outcomes. Conclusion This study offers valuable insights into the roles of TREM2, HCST, and TYROBP in tumor immunotherapy, suggesting their potential as prognostic markers and therapeutic targets for various cancers.
Collapse
Affiliation(s)
- Piao Zheng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yejun Tan
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Qing Liu
- The department of neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Changwu Wu
- The department of neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Kang
- Department of rheumatology and immunology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuzhi Liang
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan, China
| | - Kuipo Yan
- Department of cardiology, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, China
| | - Lingfeng Zeng
- Academician Workstation, Changsha Medical University, Changsha, Hunan, China
| | - Bolin Chen
- The Second Department of Thoracic Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
44
|
Fang C, Zhong R, Lu S, Yu G, Liu Z, Yan C, Gao J, Tang Y, Wang Y, Zhao Q, Feng X. TREM2 promotes macrophage polarization from M1 to M2 and suppresses osteoarthritis through the NF-κB/CXCL3 axis. Int J Biol Sci 2024; 20:1992-2007. [PMID: 38617547 PMCID: PMC11008261 DOI: 10.7150/ijbs.91519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/03/2024] [Indexed: 04/16/2024] Open
Abstract
Objective: Osteoarthritis (OA) is the most prominent chronic arthritic disease, affecting over 3 billion people globally. Synovial macrophages, as immune cells, play an essential role in cartilage damage in OA. Therefore, regulating macrophages is crucial for controlling the pathological changes in OA. Triggering receptor expressed on myeloid cells 2 (TREM2), as expressed on immune cell surfaces, such as macrophages and dendritic cells, has suppressed inflammation and regulated M2 macrophage polarization but demonstrated an unknown role in synovial macrophage polarization in OA. This study aimed to investigate TREM2 expression downregulation in OA mice macrophages. Furthermore, the expression trend of TREM2 was associated with polarization-related molecule expression in macrophages of OA mice. Results: We used TREM2 knockout (TREM2-KO) mice to observe that TREM2 deficiency significantly exacerbated the joint inflammation response in OA mice, thereby accelerating disease progression. Separating macrophages and chondrocytes from TREM2-KO mice and co-cultivating them significantly increased chondrocyte apoptosis and inhibited chondrocyte proliferation. Further, TREM2 deficiency also significantly enhanced phosphatidylinositol 3-kinase(PI3K)/AKT signaling pathway activation, increasing nuclear factor kappa light chain enhancer of activated B cells (NF-κB) signaling and C-X-C Motif Chemokine Ligand 3 (CXCL3) expression. Furthermore, NF-κB signaling pathway inhibition significantly suppressed arthritis inflammation in OA mice, thereby effectively alleviating TREM2 deficiency-related adverse effects on chondrocytes. Notably, knocking down CXCL3 of TREM2-KO mice macrophages significantly inhibits inflammatory response and promotes chondrocyte proliferation. Intravenous recombinant TREM2 protein (soluble TREM2, sTREM2) injection markedly promotes macrophage polarization from M1 to M2 and improves the joint tissue pathology and inflammatory response of OA. Conclusion: Our study reveals that TREM2 promotes macrophage polarization from M1 to M2 during OA by NF-κB/CXCL3 axis regulation, thereby improving the pathological state of OA.
Collapse
Affiliation(s)
- Chao Fang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Rui Zhong
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Shuai Lu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Gang Yu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Zhilin Liu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Chengyuan Yan
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Jingyu Gao
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Yang Tang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Yingming Wang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Qichun Zhao
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Xinzhe Feng
- Department of Joint Bone Disease Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
45
|
Creus-Muncunill J, Haure-Mirande JV, Mattei D, Bons J, Ramirez AV, Hamilton BW, Corwin C, Chowdhury S, Schilling B, Ellerby LM, Ehrlich ME. TYROBP/DAP12 knockout in Huntington's disease Q175 mice cell-autonomously decreases microglial expression of disease-associated genes and non-cell-autonomously mitigates astrogliosis and motor deterioration. J Neuroinflammation 2024; 21:66. [PMID: 38459557 PMCID: PMC10924371 DOI: 10.1186/s12974-024-03052-4] [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: 10/15/2023] [Accepted: 02/19/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an expansion of the CAG trinucleotide repeat in the Huntingtin gene (HTT). Immune activation is abundant in the striatum of HD patients. Detection of active microglia at presymptomatic stages suggests that microgliosis is a key early driver of neuronal dysfunction and degeneration. Recent studies showed that deletion of Tyrobp, a microglial protein, ameliorates neuronal dysfunction in Alzheimer's disease amyloidopathy and tauopathy mouse models while decreasing components of the complement subnetwork. OBJECTIVE While TYROBP/DAP12-mediated microglial activation is detrimental for some diseases such as peripheral nerve injury, it is beneficial for other diseases. We sought to determine whether the TYROBP network is implicated in HD and whether Tyrobp deletion impacts HD striatal function and transcriptomics. METHODS To test the hypothesis that Tyrobp deficiency would be beneficial in an HD model, we placed the Q175 HD mouse model on a Tyrobp-null background. We characterized these mice with a combination of behavioral testing, immunohistochemistry, transcriptomic and proteomic profiling. Further, we evaluated the gene signature in isolated Q175 striatal microglia, with and without Tyrobp. RESULTS Comprehensive analysis of publicly available human HD transcriptomic data revealed that the TYROBP network is overactivated in the HD putamen. The Q175 mice showed morphologic microglial activation, reduced levels of post-synaptic density-95 protein and motor deficits at 6 and 9 months of age, all of which were ameliorated on the Tyrobp-null background. Gene expression analysis revealed that lack of Tyrobp in the Q175 model does not prevent the decrease in the expression of striatal neuronal genes but reduces pro-inflammatory pathways that are specifically active in HD human brain, including genes identified as detrimental in neurodegenerative diseases, e.g. C1q and members of the Ccr5 signaling pathway. Integration of transcriptomic and proteomic data revealed that astrogliosis and complement system pathway were reduced after Tyrobp deletion, which was further validated by immunofluorescence analysis. CONCLUSIONS Our data provide molecular and functional support demonstrating that Tyrobp deletion prevents many of the abnormalities in the HD Q175 mouse model, suggesting that the Tyrobp pathway is a potential therapeutic candidate for Huntington's disease.
Collapse
Affiliation(s)
| | | | - Daniele Mattei
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Angie V Ramirez
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - B Wade Hamilton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Chuhyon Corwin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Sarah Chowdhury
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | | | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
46
|
Peng Y, Yang G, Wang S, Lin W, Zhu L, Dong W, Shen B, Nie Q, Hong S, Li L. Triggering Receptor Expressed on Myeloid Cells 2 Deficiency Exacerbates Methamphetamine-Induced Activation of Microglia and Neuroinflammation. Int J Toxicol 2024; 43:165-176. [PMID: 38006258 DOI: 10.1177/10915818231216397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2023]
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant and one of the most widely abused drugs worldwide. The continuous use of METH eventually leads to neurotoxicity and drug addiction. Studies have shown that neurotoxicity is strongly associated with METH-induced neuroinflammation, and microglia are the key drivers of neuroinflammation. Triggering receptor expressed on myeloid cells 2 (TREM2) is reported to play a key role in activation of microglia and neuroinflammation. Yet, the molecular mechanisms by which METH causes neuroinflammation and neurotoxicity remain elusive. In the current study, we investigated the role of TREM2 in neuroinflammation induced by METH in BV2 cells and the wild-type (WT) C57BL/6J mice, CX3CR1GFP/+ transgenic mice, and TREM2 knockout (KO) mice. Postmortem samples from the frontal cortex of humans with a history of METH use were also analyzed to determine the levels of TREM2, TLR4, IBA1, and IL-1β. The expression levels of TREM2, TLR4, IBA1, IL-1β, iNOS, and Arg-1 were then assessed in the BV2 cells and frontal cortex of mice and human METH users. Results revealed that the expression levels of TREM2, TLR4, IBA1, and IL-1β were significantly elevated in METH-using individuals and BV2 cells. Microglia were clearly activated in the frontal cortex of WT C57BL/6 mice and CX3CR1GFP/+ transgenic mice, and the protein levels of IBA1, TREM2, TLR4, and IL-1β were elevated in the METH-induced mouse models. Moreover, TREM2-KO mice showed further increased microglial activation, neuroinflammation, and excitotoxicity induced by METH. Thus, these findings suggest that TREM2 may be a target for regulating METH-induced neuroinflammation.
Collapse
Affiliation(s)
- Yanxia Peng
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Genmeng Yang
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Shangwen Wang
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Wanrong Lin
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihua Zhu
- Department of Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenjuan Dong
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Baoyu Shen
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Qianyun Nie
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Shijun Hong
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Lihua Li
- School of Forensic Medicine, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| |
Collapse
|
47
|
Golzari-Sorkheh M, Liyanage I, Reed MA, Weaver DF. Alzheimer's Disease and COVID-19 Pathogenic Overlap: Implications for Drug Repurposing. Can J Neurol Sci 2024; 51:161-172. [PMID: 36991574 DOI: 10.1017/cjn.2023.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
As COVID-19 continues, a safe, cost-effective treatment strategy demands continued inquiry. Chronic neuroinflammatory disorders may appear to be of little relevance in this regard; often indolent and progressive disorders characterized by neuroinflammation (such as Alzheimer's disease (AD)) are fundamentally dissimilar in etiology and symptomology to COVID-19's rapid infectivity and pathology. However, the two disorders share extensive pathognomonic features, including at membrane, cytoplasmic, and extracellular levels, culminating in analogous immunogenic destruction of their respective organ parenchyma. We hypothesize that these mechanistic similarities may extent to therapeutic targets, namely that it is conceivable an agent against AD's immunopathy may have efficacy against COVID-19 and vice versa. It is notable that while extensively investigated, no agent has yet demonstrated significant therapeutic efficacy against AD's cognitive and memory declines. Yet this very failure has driven the development of numerous agents with strong mechanistic potential and clinical characteristics. Having already approved for clinical trials, these agents may be an expedient starting point in the urgent search for an effective COVID-19 therapy. Herein, we review the overlapping Alzheimer's/ COVID-19 targets and theorize several initial platforms.
Collapse
Affiliation(s)
| | - Imindu Liyanage
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mark A Reed
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Donald F Weaver
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
48
|
Tian Y, Xiao X, Liu W, Cheng S, Qian N, Wang L, Liu Y, Ai R, Zhu X. TREM2 improves microglia function and synaptic development in autism spectrum disorders by regulating P38 MAPK signaling pathway. Mol Brain 2024; 17:12. [PMID: 38409127 PMCID: PMC10898105 DOI: 10.1186/s13041-024-01081-x] [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: 09/14/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) encompasses a diverse range of neurodevelopmental disorders, but the precise underlying pathogenesis remains elusive. This study aim to explore the potential mechanism of TREM2 in regulating microglia function in ASD. MATERIALS AND METHODS The offspring rat model of ASD was established through prenatal exposure to valproic acid (VPA), and the behavioral symptoms of the ASD model were observed. On postnatal day (PND) 7 and PND 28, the effects of prenatally exposure to VPA on synaptic development and microglia phenotype of offspring rats were observed. Primary microglia were cultured in vitro. Lentivirus and adenovirus were utilized to interfere with TREM2 and overexpress TREM2. RESULTS Prenatally VPA exposure induced offspring rats to show typical ASD core symptoms, which led to abnormal expression of synapse-related proteins in the prefrontal cortex of offspring rats, changed the phenotype of microglia in offspring rats, promoted the polarization of microglia to pro-inflammatory type, and increased inflammatory response. The experimental results in vitro showed that overexpression of TREM2 could increase the expression of Gephyrin, decrease the content of CD86 protein and increase the content of CD206 protein. In addition, after the expression of TREM2 was interfered, the content of p-P38 MAPK protein increased and the content of p-ELK-1 protein decreased. CONCLUSION The protective influence of TREM2 on the VPA-induced ASD model is attributed to its inhibition of the P38 MAPK pathway, this protective effect may be achieved by promoting the polarization of microglia to anti-inflammatory phenotype and improving the neuronal synaptic development.
Collapse
Affiliation(s)
- Yi Tian
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Xiao Xiao
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Weiliang Liu
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Shanqing Cheng
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Na Qian
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Ling Wang
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Yang Liu
- School of Pediatrics, Guizhou Medical University, Guiyang City, China
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China
| | - Rong Ai
- School of Pediatrics, Guizhou Medical University, Guiyang City, China.
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China.
| | - Xiaoping Zhu
- School of Pediatrics, Guizhou Medical University, Guiyang City, China.
- Department of Pediatrics, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, 550004, Guiyang City, China.
| |
Collapse
|
49
|
Miyake K, Ito J, Takahashi K, Nakabayashi J, Brombacher F, Shichino S, Yoshikawa S, Miyake S, Karasuyama H. Single-cell transcriptomics identifies the differentiation trajectory from inflammatory monocytes to pro-resolving macrophages in a mouse skin allergy model. Nat Commun 2024; 15:1666. [PMID: 38396021 PMCID: PMC10891131 DOI: 10.1038/s41467-024-46148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Both monocytes and macrophages are heterogeneous populations. It was traditionally understood that Ly6Chi classical (inflammatory) monocytes differentiate into pro-inflammatory Ly6Chi macrophages. Accumulating evidence has suggested that Ly6Chi classical monocytes can also differentiate into Ly6Clo pro-resolving macrophages under certain conditions, while their differentiation trajectory remains to be fully elucidated. The present study with scRNA-seq and flow cytometric analyses reveals that Ly6ChiPD-L2lo classical monocytes recruited to the allergic skin lesion sequentially differentiate into Ly6CloPD-L2hi pro-resolving macrophages, via intermediate Ly6ChiPD-L2hi macrophages but not Ly6Clo non-classical monocytes, in an IL-4 receptor-dependent manner. Along the differentiation, classical monocyte-derived macrophages display anti-inflammatory signatures followed by metabolic rewiring concordant with their ability to phagocytose apoptotic neutrophils and allergens, therefore contributing to the resolution of inflammation. The failure in the generation of these pro-resolving macrophages drives the IL-1α-mediated cycle of inflammation with abscess-like accumulation of necrotic neutrophils. Thus, we clarify the stepwise differentiation trajectory from Ly6Chi classical monocytes toward Ly6Clo pro-resolving macrophages that restrain neutrophilic aggravation of skin allergic inflammation.
Collapse
Affiliation(s)
- Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| | - Junya Ito
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazufusa Takahashi
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Jun Nakabayashi
- College of Liberal Arts and Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Frank Brombacher
- Institute of Infectious Disease and Molecular Medicine, International Center for Genetic and Biotechnology Cape Town Component & University of Cape Town, Cape Town, South Africa
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Soichiro Yoshikawa
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
50
|
Bobotis BC, Halvorson T, Carrier M, Tremblay MÈ. Established and emerging techniques for the study of microglia: visualization, depletion, and fate mapping. Front Cell Neurosci 2024; 18:1317125. [PMID: 38425429 PMCID: PMC10902073 DOI: 10.3389/fncel.2024.1317125] [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: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
The central nervous system (CNS) is an essential hub for neuronal communication. As a major component of the CNS, glial cells are vital in the maintenance and regulation of neuronal network dynamics. Research on microglia, the resident innate immune cells of the CNS, has advanced considerably in recent years, and our understanding of their diverse functions continues to grow. Microglia play critical roles in the formation and regulation of neuronal synapses, myelination, responses to injury, neurogenesis, inflammation, and many other physiological processes. In parallel with advances in microglial biology, cutting-edge techniques for the characterization of microglial properties have emerged with increasing depth and precision. Labeling tools and reporter models are important for the study of microglial morphology, ultrastructure, and dynamics, but also for microglial isolation, which is required to glean key phenotypic information through single-cell transcriptomics and other emerging approaches. Strategies for selective microglial depletion and modulation can provide novel insights into microglia-targeted treatment strategies in models of neuropsychiatric and neurodegenerative conditions, cancer, and autoimmunity. Finally, fate mapping has emerged as an important tool to answer fundamental questions about microglial biology, including their origin, migration, and proliferation throughout the lifetime of an organism. This review aims to provide a comprehensive discussion of these established and emerging techniques, with applications to the study of microglia in development, homeostasis, and CNS pathologies.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
| | - Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec City, QC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|