1
|
Yi F, Wu H, Zhao HK. Role of triggering receptor expressed on myeloid cells 1/2 in secondary injury after cerebral hemorrhage. World J Clin Cases 2025; 13:100312. [DOI: 10.12998/wjcc.v13.i9.100312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/26/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a common severe emergency in neurosurgery, causing tremendous economic pressure on families and society and devastating effects on patients both physically and psychologically, especially among patients with poor functional outcomes. ICH is often accompanied by decreased consciousness and limb dysfunction. This seriously affects patients’ ability to live independently. Although rapid advances in neurosurgery have greatly improved patient survival, there remains insufficient evidence that surgical treatment significantly improves long-term outcomes. With in-depth pathophysiological studies after ICH, increasing evidence has shown that secondary injury after ICH is related to long-term prognosis and that the key to secondary injury is various immune-mediated neuroinflammatory reactions after ICH. In basic and clinical studies of various systemic inflammatory diseases, triggering receptor expressed on myeloid cells 1/2 (TREM-1/2), and the TREM receptor family is closely related to the inflammatory response. Various inflammatory diseases can be upregulated and downregulated through receptor intervention. How the TREM receptor functions after ICH, the types of results from intervention, and whether the outcomes can improve secondary brain injury and the long-term prognosis of patients are unknown. An analysis of relevant research results from basic and clinical trials revealed that the inhibition of TREM-1 and the activation of TREM-2 can alleviate the neuroinflammatory immune response, significantly improve the long-term prognosis of neurological function in patients with cerebral hemorrhage, and thus improve the ability of patients to live independently.
Collapse
Affiliation(s)
- Fan Yi
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
| | - Hao Wu
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an 710038, Shaanxi Province, China
| | - Hai-Kang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an 710038, Shaanxi Province, China
| |
Collapse
|
2
|
Prakash P, Randolph CE, Walker KA, Chopra G. Lipids: Emerging Players of Microglial Biology. Glia 2025; 73:657-677. [PMID: 39688320 PMCID: PMC11784843 DOI: 10.1002/glia.24654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024]
Abstract
Lipids are small molecule immunomodulators that play critical roles in maintaining cellular health and function. Microglia, the resident immune cells of the central nervous system, regulate lipid metabolism both in the extracellular environment and within intracellular compartments through various mechanisms. For instance, glycerophospholipids and fatty acids interact with protein receptors on the microglial surface, such as the Triggering Receptor Expressed on Myeloid Cells 2, influencing cellular functions like phagocytosis and migration. Moreover, cholesterol is essential not only for microglial survival but, along with other lipids such as fatty acids, is crucial for the formation, function, and accumulation of lipid droplets, which modulate microglial activity in inflammatory diseases. Other lipids, including acylcarnitines and ceramides, participate in various signaling pathways within microglia. Despite the complexity of the microglial lipidome, only a few studies have investigated the effects of specific lipid classes on microglial biology. In this review, we focus on major lipid classes and their roles in modulating microglial function. We also discuss novel analytical techniques for characterizing the microglial lipidome and highlight gaps in current knowledge, suggesting new directions for future research on microglial lipid biology.
Collapse
Affiliation(s)
- Priya Prakash
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Neuroscience Institute, NYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | | | - Gaurav Chopra
- Department of ChemistryPurdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Integrative Neuroscience, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute for Drug Discovery, Purdue UniversityWest LafayetteIndianaUSA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue UniversityWest LafayetteIndianaUSA
- Regenstrief Center for Healthcare Engineering, Purdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
3
|
Wu X, Miller JA, Lee BTK, Wang Y, Ruedl C. Reducing microglial lipid load enhances β amyloid phagocytosis in an Alzheimer's disease mouse model. SCIENCE ADVANCES 2025; 11:eadq6038. [PMID: 39908361 DOI: 10.1126/sciadv.adq6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Macrophages accumulate lipid droplets (LDs) under stress and inflammatory conditions. Despite the presence of LD-loaded macrophages in many tissues, including the brain, their contribution to neurodegenerative disorders remains elusive. This study investigated the role of lipid metabolism in Alzheimer's disease (AD) by assessing the contribution of LD-loaded brain macrophages, including microglia and border-associated macrophages (BAMs), in an AD mouse model. Particularly, BAMs and activated CD11c+ microglia localized near β amyloid (Aβ) plaques exhibited a pronounced lipid-associated gene signature and a high LD load. Having observed that elevated intracellular LD content correlated inversely with microglial phagocytic activities, we subsequently inhibited LD formation specifically in CX3CR1+ brain macrophages using an inducible APP-KI/Fit2iΔMφ transgenic mouse model. We demonstrated that reducing LD content in microglia and CX3CR1+ BAMs remarkably improved their phagocytic ability. Furthermore, lowering microglial LDs consistently enhanced their efferocytosis capacities and notably reduced Aβ deposition in the brain parenchyma. Therefore, mitigating LD accumulation in brain macrophages provides perspectives for AD treatment.
Collapse
Affiliation(s)
- Xiaoting Wu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - James Alastair Miller
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bernett Teck Kwong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yulan Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
4
|
Sheng L, Gao J, Wei Q, Gong Y, Xu ZX. The glial UDP-glycosyltransferase Ugt35b regulates longevity by maintaining lipid homeostasis in Drosophila. Cell Rep 2025; 44:115099. [PMID: 39723892 DOI: 10.1016/j.celrep.2024.115099] [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: 01/29/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Lipid droplets (LDs) are dynamic organelles essential for lipid storage and organismal survival. Studies have highlighted the importance of glial function in brain LD formation during aging; however, the genes and mechanisms involved remain elusive. Here, we found that Ugt35b, a member of the uridine diphosphate (UDP)-glycosyltransferases that catalyze the transfer of glycosyl groups to acceptors, is highly expressed in glia and crucial for Drosophila lifespan. By integrating multiomics data, we demonstrated that glial Ugt35b plays key roles in regulating glycerolipid and glycerophospholipid metabolism in the brain. Notably, we found that Ugt35b and Lsd-2 are co-expressed in glia and confirmed their protein interaction in vivo. Knockdown of Ugt35b significantly reduced LD formation by downregulating Lsd-2 expression, while overexpression of Lsd-2 partially rescued the shortened lifespan in glial Ugt35b RNAi flies. Our findings reveal the crucial role of glial Ugt35b in regulating LD formation to maintain brain lipid homeostasis and support Drosophila lifespan.
Collapse
Affiliation(s)
- Lihong Sheng
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Jianpeng Gao
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qingyuan Wei
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ye Gong
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Zhi-Xiang Xu
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
5
|
He S, Xu Z, Han X. Lipidome disruption in Alzheimer's disease brain: detection, pathological mechanisms, and therapeutic implications. Mol Neurodegener 2025; 20:11. [PMID: 39871348 PMCID: PMC11773937 DOI: 10.1186/s13024-025-00803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Alzheimer's disease (AD) is among the most devastating neurodegenerative disorders with limited treatment options. Emerging evidence points to the involvement of lipid dysregulation in the development of AD. Nevertheless, the precise lipidomic landscape and the mechanistic roles of lipids in disease pathology remain poorly understood. This review aims to highlight the significance of lipidomics and lipid-targeting approaches in the diagnosis and treatment of AD. We summarized the connection between lipid dysregulation in the human brain and AD at both genetic and lipid species levels. We briefly introduced lipidomics technologies and discussed potential challenges and areas of future advancements in the lipidomics field for AD research. To elucidate the central role of lipids in converging multiple pathological aspects of AD, we reviewed the current knowledge on the interplay between lipids and major AD features, including amyloid beta, tau, and neuroinflammation. Finally, we assessed the progresses and obstacles in lipid-based therapeutics and proposed potential strategies for leveraging lipidomics in the treatment of AD.
Collapse
Affiliation(s)
- Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA
| | - Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA.
| |
Collapse
|
6
|
Zhang L, Zhou Y, Yang Z, Jiang L, Yan X, Zhu W, Shen Y, Wang B, Li J, Song J. Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J Neuroinflammation 2025; 22:7. [PMID: 39806503 PMCID: PMC11730833 DOI: 10.1186/s12974-025-03334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Lipid droplets (LDs), serving as the convergence point of energy metabolism and multiple signaling pathways, have garnered increasing attention in recent years. Different cell types within the central nervous system (CNS) can regulate energy metabolism to generate or degrade LDs in response to diverse pathological stimuli. This article provides a comprehensive review on the composition of LDs in CNS, their generation and degradation processes, their interaction mechanisms with mitochondria, the distribution among different cell types, and the roles played by these cells-particularly microglia and astrocytes-in various prevalent neurological disorders. Additionally, we also emphasize the paradoxical role of LDs in post-cerebral ischemia inflammation and explore potential underlying mechanisms, aiming to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yunfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhongbo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liangchao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyang Yan
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wenkai Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bolong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiaxi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
7
|
Navarro E, Montesinos J. Mitochondria-Associated Endoplasmic Reticulum Membranes in Microglia: One Contact Site to Rule Them all. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2025; 8:25152564241312807. [PMID: 39881949 PMCID: PMC11775980 DOI: 10.1177/25152564241312807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in maintaining tissue homeostasis by monitoring and responding to environmental changes through processes such as phagocytosis, cytokine production or synapse remodeling. Their dynamic nature and diverse functions are supported by the regulation of multiple metabolic pathways, enabling microglia to efficiently adapt to fluctuating signals. A key aspect of this regulation occurs at mitochondria-associated ER membranes (MAM), specialized contact sites between the ER and mitochondria. These structures facilitate the exchange of calcium, lipids, and metabolites and serve as metabolic and signaling hubs. This review synthesizes current research on how MAM influence microglial physiology, with an emphasis on their role in immunometabolism, offering new insights into the integration of metabolic and immune functions in the CNS and its impact in the context of neurodegeneration.
Collapse
Affiliation(s)
- Elisa Navarro
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
- Neurochemistry Research Institute, Complutense University of Madrid, Madrid, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Jorge Montesinos
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040 Madrid, Spain
| |
Collapse
|
8
|
Zhang X, Du P, Bai B, Lian X, Xue G. Molecular mechanism of METTL14-mediated m6A modification regulating microglial function post ischemic stroke. Brain Res Bull 2025; 220:111156. [PMID: 39622391 DOI: 10.1016/j.brainresbull.2024.111156] [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: 07/03/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 12/08/2024]
Abstract
This study explores the molecular mechanism of METTL14 regulating microglial function post ischemic stroke. A murine model was established by tMCAO. The neurological function was evaluated by mNSS. The cerebral infarct size and pathological changes were observed by TTC and H&E staining. M1 and M2 microglia in brain tissues were detected by flow cytometry. BV2 cells were subjected to OGD/R to establish an in vitro model. qRT-PCR and Western blot were used for detecting METTL14, PAX6, YTHDF2, TREM2, iNOS, and Arg1 expressions. The m6A level was quantitatively analyzed, and the binding of YTHDF2 or m6A to PAX6 was analyzed by RIP. PAX6 mRNA stability was assessed after actinomycin D treatment. ChIP was utilized for determining the enrichment of PAX6 on TREM2 promoter. The binding relationship between TREM2 and PAX6 was verified by dual-luciferase reporter assay. METTL14 was highly expressed after tMCAO, and silence of METTL14 alleviated symptoms of tMCAO mice and promoted microglial M2 polarization. METTL14 enhanced PAX6 mRNA m6A modification to promote YTHDF2 binding to PAX6 mRNA and its degradation. PAX6 bound to TREM2 promoter and facilitated its transcription and expression. In conclusion, METTL14-mediated m6A modification aggravates ischemic stroke by promoting microglial M1 polarization via YTHDF2/PAX6/TREM2 axis.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Pengyang Du
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Bo Bai
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xia Lian
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Guofang Xue
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
9
|
Traetta ME, Vecchiarelli HA, Tremblay MÈ. Fundamental Neurochemistry Review: Lipids across microglial states. J Neurochem 2025; 169:e16259. [PMID: 39696753 DOI: 10.1111/jnc.16259] [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: 08/08/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/20/2024]
Abstract
The capacity of immune cells to alter their function based on their metabolism is the basis of the emerging field of immunometabolism. Microglia are the resident innate immune cells of the central nervous system, and it is a current focus of the field to investigate how alterations in their metabolism impact these cells. Microglia have the ability to utilize lipids, such as fatty acids, as energy sources, but also alterations in lipids can impact microglial form and function. Recent studies highlighting different microglial states and transcriptional signatures have highlighted modifications in lipid processing as defining these states. This review highlights these recent studies and uses these altered pathways to discuss the current understanding of lipid biology in microglia. The studies highlighted here review how lipids may alter microglial phagocytic functioning or alter their pro- and anti-inflammatory balance. These studies provide a foundation by which lipid supplementation or diet alterations could influence microglial states and function. Furthermore, targets modulating microglial lipid metabolism may provide new treatment avenues.
Collapse
Affiliation(s)
- Marianela E Traetta
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Haley A Vecchiarelli
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
- Institute for Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada
- Département de médecine moléculaire, Université Laval, Québec City, Quebec, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec, Université Laval, Québec City, Quebec, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Vasilopoulou F, Piers TM, Wei J, Hardy J, Pocock JM. Amelioration of signaling deficits underlying metabolic shortfall in TREM2 R47H human iPSC-derived microglia. FEBS J 2024. [PMID: 39726135 DOI: 10.1111/febs.17353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/13/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The microglial triggering receptor expressed on myeloid cells 2 (TREM2) is required for diverse microglia responses in neurodegeneration, including immunometabolic plasticity, phagocytosis, and survival. We previously identified that patient iPSC-derived microglia (iPS-Mg) harboring the Alzheimer's disease (AD) TREM2R47H hypomorph display several functional deficits linked to metabolism. To investigate whether these deficits are associated with disruptions in metabolite signaling, we generated common variant, TREM2R47H and TREM2-/- variant human iPS-Mg. We assessed the ability of supplementation with citrate or succinate, key metabolites and cell cycle breaking points upon microglia activation, to overcome these functional deficits with potential impact on neurons. Succinate supplementation was more effective than citrate at overcoming mitochondrial deficits in OXPHOS and did not promote a glycolytic switch. Citrate enhanced the lipid content of TREM2R47H iPS-Mg and was more effective at overcoming Αβ phagocytic deficits, whereas succinate increased lipid content and phagocytic capacity in TREM2-/- iPS-Mg. Microglia cytokine secretion upon pro-inflammatory activation was moderately affected by citrate or succinate showing a condition-dependent increasing trend. Neither metabolite altered basal levels of soluble TREM2 shedding. In addition, neither citrate nor succinate enhanced glycolysis; instead, drove their effects through oxidative phosphorylation. IPS-neurons exposed to conditioned medium from TREM2 variant iPS-Mg showed changes in oxidative phosphorylation, which could be ameliorated when iPS-Mg were first treated with citrate or succinate. Our data point to discrete pathway linkage between microglial metabolism and functional outcomes with implications for AD pathogenesis and treatments.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, UK
| | - Thomas M Piers
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, UK
| | - Jingzhang Wei
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, UK
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
- NIHR University College London Hospitals Biomedical Research Centre and Institute for Advanced Study, The Hong Kong University of Science and Technology, China
| | - Jennifer M Pocock
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College London, UK
| |
Collapse
|
11
|
Li L, Sun B, Harris OA, Luo J. TGF-β Signaling in Microglia: A Key Regulator of Development, Homeostasis and Reactivity. Biomedicines 2024; 12:2468. [PMID: 39595034 PMCID: PMC11592028 DOI: 10.3390/biomedicines12112468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are crucial for normal brain development and function. They become reactive in response to brain injury and disease, a process known as microglial reactivity. This reactivity, along with microglial homeostasis, is tightly regulated by the local microenvironment and interactions with surrounding cells. The TGF-β signaling pathway plays an essential role in this regulation. Recent genetic studies employing microglia-specific manipulation of the TGF-β signaling pathway have shed light on its significance in microglial development, homeostasis and reactivity. This review provides an updated overview of how TGF-β signaling modulates microglial function and reactivity, contributing to our understanding of microglial biology in health and disease.
Collapse
Affiliation(s)
- Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Odette A. Harris
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
12
|
Wang D, Liu J, Zhu Q, Wei X, Zhang X, Chen Q, Zhao Y, Tang H, Xu W. Ouabain Ameliorates Alzheimer's Disease-Associated Neuropathology and Cognitive Impairment in FAD 4T Mice. Nutrients 2024; 16:3558. [PMID: 39458551 PMCID: PMC11510559 DOI: 10.3390/nu16203558] [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: 09/16/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Alzheimer's disease (AD) is a common clinical neurodegenerative disorder, primarily characterized by progressive cognitive decline and behavioral abnormalities. The hallmark pathological changes of AD include widespread neuronal degeneration, plaques formed by the deposition of amyloid β-protein (Aβ), and neurofibrillary tangles (NFTs). With the acceleration of global aging, the incidence of AD is rising year by year, making it a major global public health concern. Due to the complex pathology of AD, finding effective interventions has become a key focus of research. Ouabain (OUA), a cardiac glycoside, is well-known for its efficacy in treating heart disease. Recent studies have also indicated its potential in AD therapy, although its exact mechanism of action remains unclear. Methods: This study integrates bioinformatics, multi-omics technologies, and in vivo and in vitro experiments to investigate the effects of OUA on the pathophysiological changes of AD and its underlying molecular mechanisms. Results: This study analyzed the expression of the triggering receptor expressed on myeloid cells 2 (TREM2) across different stages of AD using bioinformatics. Serum samples from patients were used to validate soluble TREM2 (sTREM2) levels. Using an Aβ1-42-induced microglial cell model, we confirmed that OUA enhances the PI3K/AKT signaling pathway activation by upregulating TREM2, which reduces neuroinflammation and promotes the transition of microglia from an M1 proinflammatory state to an M2 anti-inflammatory state. To evaluate the in vivo effects of OUA, we assessed the learning and memory capacity of FAD4T transgenic mice using the Morris water maze and contextual fear conditioning tests. We used real-time quantitative PCR, immunohistochemistry, and Western blotting to measure the expression of inflammation-associated cytokines and to assess microglia polarization. OUA enhances cognitive function in FAD4T mice and has been confirmed to modulate microglial M1/M2 phenotypes both in vitro and in vivo. Furthermore, through bioinformatics analysis, molecular docking, and experimental validation, TREM2 was identified as a potential target for OUA. It regulates PI3K/Akt signaling pathway activation, playing a crucial role in OUA-mediated M2 microglial polarization and its anti-inflammatory effects in models involving Aβ1-42-stimulated BV-2 cells and FAD4T mice. Conclusions: These findings indicate that OUA exerts anti-neuroinflammatory effects by regulating microglial polarization, reducing the production of inflammatory mediators, and activating the PI3K/Akt signaling pathway. Given its natural origin and dual effects on microglial polarization and neuroinflammation, OUA emerges as a promising therapeutic candidate for neuroinflammatory diseases such as AD.
Collapse
Affiliation(s)
- Dan Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, China
| | - Jiajia Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
| | - Qizhi Zhu
- The Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China; (Q.Z.); (X.W.)
| | - Xin Wei
- The Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China; (Q.Z.); (X.W.)
| | - Xiang Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
| | - Qi Chen
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
| | - Yu Zhao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
| | - Heng Tang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
| | - Weiping Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (D.W.); (J.L.); (X.Z.); (Q.C.); (Y.Z.); (H.T.)
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, China
| |
Collapse
|
13
|
Liu C, Zhou X. TREM2 Impairs Glycolysis to Interrupt Microglial M1 Polarization and Inflammation via JAK2/STAT3 Axis. Cell Biochem Biophys 2024:10.1007/s12013-024-01520-5. [PMID: 39240442 DOI: 10.1007/s12013-024-01520-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Cerebral ischemia/reperfusion injury (IRI) is a primary pathophysiological basis of ischemic stroke, a dreadful cerebrovascular event carrying substantial disability and lethality. Triggering receptor expressed on myeloid cells 2 (TREM2) is a membrane glycoprotein that has been notified as a protective factor for cerebral ischemic stroke. On this basis, the paper is thereby goaled to interpret the probable activity and downstream mechanism of TREM2 against cerebral IRI. Cerebral IRI was simulated in murine microglial BV2 cells under oxygen-glucose deprivation and reperfusion (OGD/R) conditions. Western blotting ascertained the expressions of TREM2 and janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) axis-associated proteins. ELISA and RT-qPCR assayed the secretion of inflammatory cytokines. Immunofluorescence and western blotting estimated macrophage polarization. Glycolysis activation was measured through evaluating lactic acid and extracellular acidification rate (ECAR). RT-qPCR and western blotting examined the expressions of glycolytic genes. TREM2 was abnormally expressed and JAK2/STAT3 axis was aberrantly activated in BV2 cells in response to OGD/R. Elevation of TREM2 repressed the inflammatory reaction and glycolysis, inhibited the JAK2/STAT3 axis, whereas promoted M1-to-M2 polarization in OGD/R-injured BV2 cells. Upregulated TREM2 inactivated the glycolytic pathway to relieve OGD/R-induced inflammatory injury and M1 macrophage polarization. Besides, STAT3 activator, colivelin, aggravated the glycolysis, inflammatory injury and drove M1-like macrophage polarization in TREM2-overexpressing BV2 cells exposed to OGD/R. Collectively, TREM2 might produce anti-inflammatory potential in cerebral IRI, which might dependent on the inactivation of glycolytic pathway via intermediating the JAK2/STAT3 axis.
Collapse
Affiliation(s)
- Chanyuan Liu
- Psychiatric Ward 1, Wuhan Wuchang Hospital, Wuhan, 430061, Hubei, China
| | - Xueying Zhou
- Department of Psychiatry, Liyuan Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430077, Hubei, China.
| |
Collapse
|
14
|
Li ZR, Wang YY, Wang ZH, Qin QL, Huang C, Shi GS, He HY, Deng YH, He XY, Zhao XM. The positive role of transforming growth factor-β1 in ischemic stroke. Cell Signal 2024; 121:111301. [PMID: 39019338 DOI: 10.1016/j.cellsig.2024.111301] [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: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Ischemic stroke is one of the most disabling and fatal diseases around the world. The damaged brain tissues will undergo excessive autophagy, vascular endothelial cells injury, blood-brain barrier (BBB) impairment and neuroinflammation after ischemic stroke. However, there is no unified viewpoint on the underlying mechanism of brain damage. Transforming growth factor-β1 (TGF-β1), as a multi-functional cytokine, plays a crucial role in the intricate pathological processes and helps maintain the physiological homeostasis of brain tissues through various signaling pathways after ischemic stroke. In this review, we summarize the protective role of TGF-β1 in autophagic flux, BBB, vascular remodeling, neuroinflammation and other aspects after ischemic stroke. Based on the review, we believe that TGF-β1 could serve as a key target for treating ischemic stroke.
Collapse
Affiliation(s)
- Zi-Rong Li
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Yong-Yan Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Zi-Han Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Qi-Lin Qin
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Cheng Huang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Guang-Sen Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Hong-Yun He
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| | - Yi-Hao Deng
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Xiu-Ying He
- Department of Anesthesiology, Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao-Ming Zhao
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
15
|
Winer BY, Settle AH, Yakimov AM, Jeronimo C, Lazarov T, Tipping M, Saoi M, Sawh A, Sepp ALL, Galiano M, Perry JSA, Wong YY, Geissmann F, Cross J, Zhou T, Kam LC, Pasolli HA, Hohl T, Cyster JG, Weiner OD, Huse M. Plasma membrane abundance dictates phagocytic capacity and functional cross-talk in myeloid cells. Sci Immunol 2024; 9:eadl2388. [PMID: 38848343 PMCID: PMC11485225 DOI: 10.1126/sciimmunol.adl2388] [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: 10/06/2023] [Accepted: 05/15/2024] [Indexed: 06/09/2024]
Abstract
Professional phagocytes like neutrophils and macrophages tightly control what they consume, how much they consume, and when they move after cargo uptake. We show that plasma membrane abundance is a key arbiter of these cellular behaviors. Neutrophils and macrophages lacking the G protein subunit Gβ4 exhibited profound plasma membrane expansion, accompanied by marked reduction in plasma membrane tension. These biophysical changes promoted the phagocytosis of bacteria, fungus, apoptotic corpses, and cancer cells. We also found that Gβ4-deficient neutrophils are defective in the normal inhibition of migration following cargo uptake. Sphingolipid synthesis played a central role in these phenotypes by driving plasma membrane accumulation in cells lacking Gβ4. In Gβ4 knockout mice, neutrophils not only exhibited enhanced phagocytosis of inhaled fungal conidia in the lung but also increased trafficking of engulfed pathogens to other organs. Together, these results reveal an unexpected, biophysical control mechanism central to myeloid functional decision-making.
Collapse
Affiliation(s)
- Benjamin Y. Winer
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Alexander H. Settle
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Carlos Jeronimo
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Murray Tipping
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Michelle Saoi
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | | | - Anna-Liisa L. Sepp
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - Michael Galiano
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin S. A. Perry
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Yung Yu Wong
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Frederic Geissmann
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Justin Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Ting Zhou
- Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center; New York, NY, USA
- The SKI Stem Cell Research Facility, The Center for Stem Cell Biology and Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Lance C. Kam
- Department of Biomedical Engineering, Columbia University; New York, NY, USA
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University; New York, NY, USA
| | - Tobias Hohl
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, CA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Orion D. Weiner
- Cardiovascular Research Institute, University of California San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco; San Francisco, CA, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center; New York, NY, USA
| |
Collapse
|
16
|
Li M, Wang M, Zhao Y, Zhong R, Chen W, Lei X, Wu X, Han J, Lei L, Wang Q, Luo G, Wei M. Effects of elevated remnant cholesterol on outcomes of acute ischemic stroke patients receiving mechanical thrombectomy. J Thromb Thrombolysis 2024; 57:390-401. [PMID: 38180591 DOI: 10.1007/s11239-023-02939-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2023] [Indexed: 01/06/2024]
Abstract
OBJECTIVE Large cohort studies provided evidence that elevated remnant cholesterol (RC) was an important risk factor for ischemic stroke. However, the association between high RC and clinical outcomes in acute ischemic stroke (AIS) individuals was still undetermined. METHODS This retrospective study enrolled 165 AIS patients undergoing mechanical thrombectomy in one tertiary stroke center. We divided patients into two groups based on the median of their RC levels (0.49 mmol/L). The modified Rankin Scale (mRS) was used to evaluate the primary outcome 90 days after the onset of symptoms. The mRS scores ≤ 2 and ≤ 1 at 90 days were deemed as favorable and excellent outcomes, respectively. RESULTS In the overall AIS patients undergoing mechanical thrombectomy, there was no obvious distinction between the high and low RC group at 90-day favorable outcome (41.0% vs. 47.1%, P = 0.431) or excellent outcome (23.1% vs. 31.0%, P = 0.252). In the subgroup analysis stratified by stroke etiology, non-large artery atherosclerosis (non-LAA) stroke patients yielded with less favorable or excellent prognosis in the high RC group (26.8% vs. 46.8%, adjusted OR = 0.31, 95%CI: 0.11-0.85, P = 0.023; or 12.2% vs. 29.0%, adjusted OR = 0.18, 95%CI: 0.04-0.80, P = 0.024, respectively.). Post hoc power analyses indicated that the power was sufficient for favorable outcome (80.38%) and excellent outcome (88.72%) in non-LAA stroke patients. Additionally, RC can enhance the risk prediction value of a poor outcome (mRS scores 3-6) based on traditional risk indicators (including age, initial NIHSS score, operative duration, and neutrophil-to-lymphocyte ratio) for non-LAA stroke patients (AUC = 0.86, 95%CI: 0.79-0.94, P < 0.001). CONCLUSION In AIS patients undergoing mechanical thrombectomy, elevated RC was independently related to poor outcome for non-LAA stroke patients, but not to short-term prognosis of LAA stroke patients.
Collapse
Affiliation(s)
- Mengmeng Li
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Neurology, Xi'an Children's Hospital, Xi'an, China
| | - Yixin Zhao
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Zhong
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wanying Chen
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiangyu Lei
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyu Wu
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaxin Han
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Lei
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qingfan Wang
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guogang Luo
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Meng Wei
- Stroke Centre, Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|