1
|
Zhang Y, Shi Y, Wang L, Li Z, Wang Y, Yan J, Sun X, Luo Q, Li L. TREM2 activation reduces white matter injury via PI3K/Akt/GSK-3β signalling after intracerebral haemorrhage. Br J Pharmacol 2025; 182:2542-2559. [PMID: 39965632 DOI: 10.1111/bph.17475] [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: 09/07/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND White matter injury (WMI) considerably exacerbates the prognosis following intracerebral haemorrhage (ICH). While the triggering receptor on myeloid cells 2 (TREM2) is recognized for its neuroprotective roles in a range of neurological disorders through the modulation of neuroinflammation, phagocytosis, promoting cell survival, its specific function in WMI after ICH has yet to be fully elucidated. METHODS This study involved inducing ICH in mice through autologous blood injection. Neurological functions were tested via behavioural assessments and electrophysiological recordings. WMI was examined using immunofluorescence, Luxol fast blue staining, MRI and transmission electron microscopy. Microglia were isolated and analysed using real-time polymerase chain reaction (PCR). Microglia depletion was achieved with PLX3397, primary cultures of microglia and oligodendrocytes were investigated. RESULTS The activation of TREM2 resulted in improved neurological outcomes after ICH, correlated with reduced WMI, demonstrated by decreased white matter loss in the corpus striatum, reduced damage to the nodes of Ranvier, and better preservation of myelin and white matter tract integrity. These neuroprotective effects were attributed to changes in microglial states mediated via the PI3K/Akt/GSK-3β signalling pathway. However, the neuroprotective advantages conferred by TREM2 activation were negated in TREM2 KO mice, either through microglia depletion or inhibition of PI3K. CONCLUSIONS This research is the first to illustrate that TREM2 activation mitigates WMI following ICH through a microglia-dependent mechanism involving the PI3K/Akt/GSK-3β pathway. TREM2 represents a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, China
| | - You Shi
- Department of Neuro-oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Wang
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, China
| | - Zhao Li
- Emergency Department, Chengdu First People's Hospital, Chengdu, China
| | - Yingwen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Yan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Luo
- Department of Ultrasound, Chong Gang General Hospital, Chongqing, China
| | - Lin Li
- Department of Neurosurgery, chong gang general hospital, Chongqing, China
| |
Collapse
|
2
|
Sun Z, Wang X, Shi C, Yu T, Xu W, Ji X, Su K, Yan H, Shan Y, Xie T, Xu J, Zhao X, Shan J. TREM2 modulates lipid metabolism to alleviate airway inflammation in asthma: A potential therapeutic target. Int J Biol Macromol 2025; 308:142306. [PMID: 40154695 DOI: 10.1016/j.ijbiomac.2025.142306] [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: 10/31/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Asthma is a chronic inflammatory disease characterized by airway hyperresponsiveness (AHR) and immune cell infiltration. TREM2 (Triggering Receptor Expressed on Myeloid cells 2), known for its role in lipid metabolism and inflammation, was found to be upregulated in asthma. Using Trem2-/- mice, we observed that TREM2 deletion significantly reduces airway inflammation and AHR. This effect is achieved through the modulation of triglyceride (TG) metabolism, leading to increased TG synthesis, decreased lipolysis, and reduced release of pro-inflammatory free fatty acids (FFAs), particularly arachidonic acid. The study reveals a novel role for TREM2 in regulating lipid metabolism in asthma, suggesting that targeting TREM2 may offer new therapeutic opportunities for managing asthma and related inflammatory conditions.
Collapse
Affiliation(s)
- Zhengpeng Sun
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Wang
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chen Shi
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tao Yu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weichen Xu
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xinyu Ji
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ke Su
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Hua Yan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yiwen Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tong Xie
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jianya Xu
- Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xia Zhao
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Jinjun Shan
- Jiangsu Key Laboratory of Children's Health and Chinese Medicine, State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Zhang L, Xiang X, Li Y, Bu G, Chen XF. TREM2 and sTREM2 in Alzheimer's disease: from mechanisms to therapies. Mol Neurodegener 2025; 20:43. [PMID: 40247363 PMCID: PMC12004684 DOI: 10.1186/s13024-025-00834-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: 12/12/2024] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor predominantly expressed by microglia in the brain. Recent studies have established TREM2 as a central immune signaling hub in neurodegeneration, where it triggers immune responses upon sensing pathological development and tissue damages. TREM2 binds diverse ligands and activates downstream pathways that regulate microglial phagocytosis, inflammatory responses, and metabolic reprogramming. Interestingly, TREM2 exists both in its membrane-bound form and as a soluble variant (sTREM2), that latter is generated through proteolytic shedding or alternative splicing and can be detected in cerebrospinal fluid and plasma. Emerging clinical and preclinical evidence underscores the potential of TREM2 and sTREM2 as diagnostic biomarkers and therapeutic targets in Alzheimer's disease (AD). This review provides a comprehensive overview of the molecular functions, regulatory mechanisms, and pathological implications of TREM2 and sTREM2 in AD. Furthermore, we explore their potential roles in diagnostics and therapeutics while suggesting key research directions for advancing TREM2/sTREM2-based strategies in combating AD.
Collapse
Affiliation(s)
- Lianshuai Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Xianyuan Xiang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518055, China.
| | - Yahui Li
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Guojun Bu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiao-Fen Chen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
4
|
Zhang Y, Zhang S, Zhao X, Wu P, Ying Y, Wu L, Zhuang J, Chen Z, Chao Y, Dong X, Zhao RC, Wang J. ATP11B Modulates Microglial Lipid Metabolism and Alleviates Alzheimer's Disease Pathology. MedComm (Beijing) 2025; 6:e70139. [PMID: 40123832 PMCID: PMC11928880 DOI: 10.1002/mco2.70139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 11/28/2024] [Accepted: 12/21/2024] [Indexed: 03/25/2025] Open
Abstract
Abnormal lipid metabolism in microglia leads to the formation of pathological lipid droplets (LDs), a phenomenon also observed in neurodegenerative diseases such as Alzheimer's disease (AD). The abnormal accumulation of LDs disrupts normal cellular function and exacerbates the pathological process of AD. ATP11B is a P4-ATPase and the expression of Atp11b changes in the brain of patients with AD and diseases of lipid metabolism. The present study aimed to explore the regulatory role of ATP11B in microglial lipid metabolism and assess the potential of ATP11B as a therapeutic target for AD. Atp11b deficiency caused excessive fatty acid uptake and activated the PPAR signaling pathway, resulting in abnormal synthesis of neutral lipids and mitochondrial energy metabolism in microglia. Further results showed that Atp11b deficiency led to the accumulation of pathological LDs in microglia and AD mice. Conversely, overexpression of Atp11b alleviated exploratory behavior impairment, learning and memory impairment, LD accumulation, beta-amyloid (Aβ) deposition, and inflammatory response in the brain of AD mice. These findings provide important clues for a better understanding of the pathogenesis of AD and for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Life SciencesShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Shibo Zhang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Xuyu Zhao
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Peiru Wu
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Yiwei Ying
- School of Life SciencesShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Lingling Wu
- School of Life SciencesShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Junyi Zhuang
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Zixin Chen
- School of Life SciencesShanghai UniversityShanghaiChina
| | - Yufan Chao
- School of MedicineShanghai UniversityShanghaiChina
| | - Xin Dong
- School of MedicineShanghai UniversityShanghaiChina
| | - Robert Chunhua Zhao
- School of Life SciencesShanghai UniversityShanghaiChina
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Centre of Excellence in Tissue EngineeringChinese Academy of Medical SciencesBeijingChina
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381)BeijingChina
| | - Jiao Wang
- School of Life SciencesShanghai UniversityShanghaiChina
| |
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
|
Mao Y, Wan J, Lin B, Xu P, Zhang K, Jin M, Xuan S, Wang M, Du J, Zhang L, Tang Z. Felodipine Promotes the Recovery of Mice With Spinal Cord Injury by Activating Macrolipophagy Through the AMPK-mTOR Pathway. J Cell Mol Med 2025; 29:e70543. [PMID: 40259510 PMCID: PMC12011640 DOI: 10.1111/jcmm.70543] [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: 10/28/2024] [Revised: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025] Open
Abstract
Spinal cord injury (SCI) is a serious clinical condition characterised by extensive mechanical damage that compromises the tissue structure and microenvironment of the affected area. This damage leads to the formation of fibrotic blood vessels and impaired energy metabolism, both of which hinder recovery. Felodipine, a clinically approved antihypertensive drug, acts as a selective calcium antagonist, primarily inhibiting extracellular calcium influx in arteriolar smooth muscle and selectively dilating arterioles. Additionally, felodipine has been demonstrated to induce autophagy. Considering these properties collectively, we hypothesised that felodipine could modulate the microenvironment of the injured spinal cord. In this study, we employed immunofluorescence and Western blot analyses to evaluate the effects of felodipine on microenvironment repair and neuroprotection, both in vitro and in vivo. Particular attention was given to its regulatory role in AMPK-mTOR pathway-mediated macrolipophagy. Our results demonstrated that felodipine effectively improved the injured spinal cord microenvironment by activating macrolipophagy, facilitating the clearance of myelin debris. Furthermore, felodipine promoted the restoration of endothelial cell tight junctions, thereby enhancing the integrity of the blood-spinal cord barrier. This attenuation of barrier disruption after SCI contributed to improved neuronal survival. These findings expanded the clinical application prospect of felodipine and presented new therapeutic avenues for treating SCI.
Collapse
Affiliation(s)
- Yuqin Mao
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Jinlong Wan
- Department of GastroenterologyGaozhou People's HospitalMaomingChina
| | - Binghao Lin
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Pengtao Xu
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Ke Zhang
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Mengyun Jin
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Shaoyan Xuan
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Minxiu Wang
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Jiqing Du
- School of Life and Health TechnologyDongguan University of TechnologyDongguanChina
| | - Lin Zhang
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| | - Zhihua Tang
- Department of PharmacyShaoxing People's HospitalShaoxingChina
| |
Collapse
|
7
|
Yu Z, Shen Y, Zhang H, Zhang W, Zhang X, Wang Y, Nie C, Zhou J, Gao A, Liang H. Exploring the genetic causal inference between plasma lipidome and hemorrhagic stroke. J Stroke Cerebrovasc Dis 2025; 34:108252. [PMID: 39875008 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108252] [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: 06/15/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Recent research indicates that the plasma lipidome composition may undergo alterations following hemorrhagic stroke. Nevertheless, the causal inference between plasma lipidome and hemorrhagic stroke remains elusive. MATERIALS AND METHODS Exposure data were achieved from a recent Genome-wide Association Study (GWAS) study of 179 lipid species involving 7174 individuals, while the outcome data were obtained from the FinnGen consortium (R10), including intracerebral hemorrhage (ICH), subarachnoid hemorrhage (SAH), and non-traumatic intracranial hemorrhage (nITH). Two-sample bidirectional Mendelian randomization (MR) analyses were used to assess the causal inference between lipidome and hemorrhagic stroke, and inverse variance weighted served as the main method. Genetic correlations between lipidome and hemorrhagic stroke were assessed using linkage disequilibrium score regression (LDSC). RESULTS After the false discovery rate (FDR) correction, Phosphatidylcholine (O-18:2_20:4) was identified as a substantial risk factor for ICH (OR,1.199; 95 % CI, 1.073-1.341; PFDR = 0.073). Alternatively, Phosphatidylinositol (16:0_18:1) was a relevant protective factor (OR, 0.773; 95 % CI, 0.666-0.896; PFDR = 0.069). Furthermore, the Sterol ester (27:1/20:3) (OR, 1.138; 95 % CI, 1.024-1.264; PFDR = 0.086) was identified as the prominent risk factor for SAH. Finally, Sterol ester (27:1/20:4) (OR, 1.073; 95 % CI, 1.026-1.121; PFDR = 0.030) and Phosphatidylinositol (16:0_18:1) levels (OR, 0.794; 95 % CI, 0.709-0.889; PFDR = 0.007) was identified as risk and protective factors for nITH, respectively. CONCLUSIONS The causal relationship between plasma lipidome and hemorrhagic stroke is evident. Studying the plasma lipidome offers promising preventive strategies and potential therapeutic approaches for hemorrhagic stroke.
Collapse
Affiliation(s)
- Zhao Yu
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Yingjie Shen
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Haopeng Zhang
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Wei Zhang
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xi Zhang
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Yaolou Wang
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Chenyi Nie
- School of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Jiaxin Zhou
- School of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Aili Gao
- School of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China.
| | - Hongsheng Liang
- NHC Key Laboratory of Cell Transplantation, Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
8
|
Zhang X, Li H, Gu Y, Ping A, Chen J, Zhang Q, Xu Z, Wang J, Tang S, Wang R, Lu J, Lu L, Jin C, Jin Z, Zhang J, Shi L. Repair-associated macrophages increase after early-phase microglia attenuation to promote ischemic stroke recovery. Nat Commun 2025; 16:3089. [PMID: 40164598 PMCID: PMC11958652 DOI: 10.1038/s41467-025-58254-y] [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: 06/12/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Ischemic stroke recovery involves dynamic interactions between the central nervous system and infiltrating immune cells. Peripheral immune cells compete with resident microglia for spatial niches in the brain, but how modulating this balance affects recovery remains unclear. Here, we use PLX5622 to create spatial niches for peripheral immune cells, altering the competition between infiltrating immune cells and resident microglia in male mice following transient middle cerebral artery occlusion (tMCAO). We find that early-phase microglia attenuation promotes long-term functional recovery. This intervention amplifies a subset of monocyte-derived macrophages (RAMf) with reparative properties, characterized by high expression of GPNMB and CD63, enhanced lipid metabolism, and pro-angiogenic activity. Transplantation of RAMf into stroke-affected mice improves white matter integrity and vascular repair. We identify Mafb as the transcription factor regulating the reparative phenotype of RAMf. These findings highlight strategies to optimize immune cell dynamics for post-stroke rehabilitation.
Collapse
Affiliation(s)
- Xiaotao Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Huaming Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Yichen Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - An Ping
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jiarui Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Qia Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhouhan Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Junjie Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Shenjie Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Rui Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Lingxiao Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Chenghao Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Ziyang Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Montilla A, Zabala A, Calvo I, Bosch-Juan M, Tomé-Velasco I, Mata P, Koster M, Sierra A, Kooistra SM, Soria FN, Eggen BJL, Fresnedo O, Fernández JA, Tepavcevic V, Matute C, Domercq M. Microglia regulate myelin clearance and cholesterol metabolism after demyelination via interferon regulatory factor 5. Cell Mol Life Sci 2025; 82:131. [PMID: 40137979 PMCID: PMC11947375 DOI: 10.1007/s00018-025-05648-2] [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: 07/05/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025]
Abstract
Interferon regulatory factor 5 (IRF5) is a transcription factor that plays a role in orchestrating innate immune responses, particularly in response to viral infections. Notably, IRF5 has been identified as a microglia risk gene linked to multiple sclerosis (MS), but its specific role in MS pathogenesis remains unclear. Through the use of Irf5-/- mice, our study uncovers a non-canonical function of IRF5 in MS recovery. Irf5-/- mice exhibited increased damage in an experimental autoimmune encephalomyelitis (EAE) model and demonstrated impaired oligodendrocyte recruitment into the lesion core following lysolecithin-induced demyelination. Transcriptomic and lipidomic analyses revealed that IRF5 has a role in microglia-mediated myelin phagocytosis, lipid metabolism, and cholesterol homeostasis. Indeed, Irf5-/- microglia phagocytose myelin, but myelin debris is not adequately degraded, leading to an accumulation of lipid droplets, cholesterol esters, and cholesterol crystals within demyelinating lesions. This abnormal buildup can hinder remyelination processes. Importantly, treatments that promote cholesterol transport were found to reduce lipid droplet accumulation and mitigate the exacerbated damage in Irf5-/- mice with EAE. Altogether, our study identified the antiviral transcription factor IRF5 as a key transcriptional regulator of lipid degradation and cholesterol homeostasis and suggest that loss of IRF5 function leads to pathogenic lipid accumulation in microglia, thereby obstructing remyelination. These data and the fact that Irf5 polymorphisms are significantly associated with MS, highlight IRF5 as a potential therapeutic target to promote regenerative responses.
Collapse
Affiliation(s)
- Alejandro Montilla
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - Alazne Zabala
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Ibai Calvo
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Marina Bosch-Juan
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Irene Tomé-Velasco
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Paloma Mata
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Mirjam Koster
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Federico N Soria
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Olatz Fresnedo
- Lipids & Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - José Andrés Fernández
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Vanja Tepavcevic
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - María Domercq
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
| |
Collapse
|
10
|
Takalo M, Jeskanen H, Rolova T, Kervinen I, Hellén M, Heikkinen S, Koivisto H, Jokivarsi K, Müller SA, Koivumäki EM, Mäkinen P, Juopperi SP, Willman RM, Sinisalo R, Hoffmann D, Jäntti H, Peitz M, Fließbach K, Kuulasmaa T, Natunen T, Kemppainen S, Poutiainen P, Leinonen V, Malm T, Martiskainen H, Ramirez A, Haapasalo A, Lichtenthaler SF, Tanila H, Haass C, Rinne J, Koistinaho J, Hiltunen M. The protective PLCγ2-P522R variant mitigates Alzheimer's disease-associated pathologies by enhancing beneficial microglial functions. J Neuroinflammation 2025; 22:64. [PMID: 40038760 PMCID: PMC11881468 DOI: 10.1186/s12974-025-03387-6] [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: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Phospholipase C gamma 2, proline 522 to arginine (PLCγ2-P522R) is a protective variant that reduces the risk of Alzheimer's disease (AD). Recently, it was shown to mitigate β-amyloid pathology in a 5XFAD mouse model of AD. Here, we investigated the protective functions of the PLCγ2-P522R variant in a less aggressive APP/PS1 mouse model of AD and assessed the underlying cellular mechanisms using mouse and human microglial models. METHODS The effects of the protective PLCγ2-P522R variant on microglial activation, AD-associated β-amyloid and neuronal pathologies, and behavioral changes were investigated in PLCγ2-P522R knock-in variant mice crossbred with APP/PS1 mice. Transcriptomic, proteomic, and functional studies were carried out using microglia isolated from mice carrying the PLCγ2-P522R variant. Finally, microglia-like cell models generated from human blood and skin biopsy samples of PLCγ2-P522R variant carriers were employed. RESULTS The PLCγ2-P522R variant decreased β-amyloid plaque count and coverage in female APP/PS1 mice. Moreover, the PLCγ2-P522R variant promoted anxiety in these mice. The area of the microglia around β-amyloid plaques was also increased in mice carrying the PLCγ2-P522R variant, while β-amyloid plaque-associated neuronal dystrophy and the levels of certain cytokines, including IL-6 and IL-1β, were reduced. These alterations were revealed through [18F]FEPPA PET imaging and behavioral studies, as well as various cytokine immunoassays, transcriptomic and proteomic analyses, and immunohistochemical analyses using mouse brain tissues. In cultured mouse primary microglia, the PLCγ2-P522R variant reduced the size of lipid droplets. Furthermore, transcriptomic and proteomic analyses revealed that the PLCγ2-P522R variant regulated key targets and pathways involved in lipid metabolism, mitochondrial fatty acid oxidation, and inflammatory/interferon signaling in acutely isolated adult mouse microglia and human monocyte-derived microglia-like cells. Finally, the PLCγ2-P522R variant also increased mitochondrial respiration in human iPSC-derived microglia. CONCLUSIONS These findings suggest that the PLCγ2-P522R variant exerts protective effects against β-amyloid and neuronal pathologies by increasing microglial responsiveness to β-amyloid plaques in APP/PS1 mice. The changes observed in lipid/fatty acid and mitochondrial metabolism revealed by the omics and metabolic assessments of mouse and human microglial models suggest that the protective effects of the PLCγ2-P522R variant are potentially associated with increased metabolic capacity of microglia.
Collapse
Affiliation(s)
- Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| | - Heli Jeskanen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Taisia Rolova
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Inka Kervinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Marianna Hellén
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Hennariikka Koivisto
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kimmo Jokivarsi
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Esa-Mikko Koivumäki
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Petra Mäkinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | | | - Rosa Sinisalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Dorit Hoffmann
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Jäntti
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Bonn, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Klaus Fließbach
- Department of Old Age Psychiatry and Cognitive Disorders, University of Bonn Medical Center, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
| | - Teemu Kuulasmaa
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Teemu Natunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Susanna Kemppainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Pekka Poutiainen
- Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Ville Leinonen
- Department of Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henna Martiskainen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Disease (CECAD), University of Cologne, Cologne, Germany
| | - Annakaisa Haapasalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Neuroproteomics, School of Medicine and Health, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Heikki Tanila
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Juha Rinne
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Jari Koistinaho
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
11
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [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] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
12
|
Zhang Y, Chen Y, Zhuang C, Qi J, Zhao RC, Wang J. Lipid droplets in the nervous system: involvement in cell metabolic homeostasis. Neural Regen Res 2025; 20:740-750. [PMID: 38886939 PMCID: PMC11433920 DOI: 10.4103/nrr.nrr-d-23-01401] [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: 08/19/2023] [Revised: 12/05/2023] [Accepted: 01/20/2024] [Indexed: 06/20/2024] Open
Abstract
Lipid droplets serve as primary storage organelles for neutral lipids in neurons, glial cells, and other cells in the nervous system. Lipid droplet formation begins with the synthesis of neutral lipids in the endoplasmic reticulum. Previously, lipid droplets were recognized for their role in maintaining lipid metabolism and energy homeostasis; however, recent research has shown that lipid droplets are highly adaptive organelles with diverse functions in the nervous system. In addition to their role in regulating cell metabolism, lipid droplets play a protective role in various cellular stress responses. Furthermore, lipid droplets exhibit specific functions in neurons and glial cells. Dysregulation of lipid droplet formation leads to cellular dysfunction, metabolic abnormalities, and nervous system diseases. This review aims to provide an overview of the role of lipid droplets in the nervous system, covering topics such as biogenesis, cellular specificity, and functions. Additionally, it will explore the association between lipid droplets and neurodegenerative disorders. Understanding the involvement of lipid droplets in cell metabolic homeostasis related to the nervous system is crucial to determine the underlying causes and in exploring potential therapeutic approaches for these diseases.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yiqing Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Cheng Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy, Beijing, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
13
|
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
|
14
|
Samuels JD, Lukens JR. Myelin debris as an initiator of microglial dysfunction and neuropathology in Alzheimer's disease. Cell Mol Immunol 2025; 22:208-210. [PMID: 39809887 PMCID: PMC11782469 DOI: 10.1038/s41423-024-01243-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Affiliation(s)
- Joshua D Samuels
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA, 22908, USA.
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22908, USA.
- Immunology Training Program, University of Virginia, Charlottesville, VA, 22908, USA.
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA, 22908, USA.
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22908, USA.
- Immunology Training Program, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
15
|
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
|
16
|
Tang Y, Zhang L, Huang P, She Z, Luo S, Peng H, Chen Y, Luo J, Duan W, Xiao Y, Liu L, Liu L. Understanding the intricacies of cellular mechanisms in remyelination: The role of circadian rhythm. Neurochem Int 2025; 183:105929. [PMID: 39756585 DOI: 10.1016/j.neuint.2025.105929] [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: 09/20/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The term "circadian rhythm" refers to the 24-h oscillations found in various physiological processes in organisms, responsible for maintaining bodily homeostasis. Many neurological diseases mainly involve the process of demyelination, and remyelination is crucial for the treatment of neurological diseases. Current research mainly focuses on the key role of circadian clocks in the pathophysiological mechanisms of multiple sclerosis. Various studies have shown that the circadian rhythm regulates various cellular molecular mechanisms and signaling pathways involved in remyelination. The process of remyelination is primarily mediated by oligodendrocyte precursor cells (OPCs), oligodendrocytes, microglia, and astrocytes. OPCs are activated, proliferate, migrate, and ultimately differentiate into oligodendrocytes after demyelination, involving many key signaling pathway and regulatory factors. Activated microglia secretes important cytokines and chemokines, promoting OPC proliferation and differentiation, and phagocytoses myelin debris that inhibits remyelination. Astrocytes play a crucial role in supporting remyelination by secreting signals that promote remyelination or facilitate the phagocytosis of myelin debris by microglia. Additionally, cell-to-cell communication via gap junctions allows for intimate contact between astrocytes and oligodendrocytes, providing metabolic support for oligodendrocytes. Therefore, gaining a deeper understanding of the mechanisms and molecular pathways of the circadian rhythm at various stages of remyelination can help elucidate the fundamental characteristics of remyelination and provide insights into treating demyelinating disorders.
Collapse
Affiliation(s)
- Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yuqiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Jinwen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Wangxin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| |
Collapse
|
17
|
Guo D, Wu J, Shen C, Zhang A, Zou T, Chen K, Huang W, Pan Y, Shen Y, Ji P, Zhong Y, Wen Q, Kong B, Xiang M, Ye B. Upregulation of LXRβ/ABCA1 pathway alleviates cochlear hair cell senescence of C57BL/6 J mice via reducing lipid droplet accumulation. Biogerontology 2025; 26:49. [PMID: 39890652 DOI: 10.1007/s10522-025-10192-4] [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: 11/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Senescence and loss of cochlear hair cells is an important pathologic basis of age-related hearing loss. Lipid droplet accumulation has previously been shown to play an important role in neurodegeneration; however, its role in age-related hearing loss has not yet been investigated. LXRβ/ABCA1 is a key pathway that regulates lipid metabolism, while its dysfunction can cause abnormal accumulation of lipid droplets in neurons, leading to neurodegeneration. In this study, we found that decreased expression of LXRβ/ABCA1, elevated levels of lipid droplet accumulation, and increased activation of the NLRP3 inflammasome were demonstrated in senescent cochlear hair cells in both animal and cellular models of age-related hearing loss. We then manipulated the LXRβ/ABCA1 pathway transduction of cochlear hair cells. Upregulation of LXRβ/ABCA1 in senescent hair cells was found to reduce the accumulation of lipid droplets, inhibit NLRP3 inflammasome activation, and ultimately alleviate cochlear hair cell senescence. In our study, we also found that NLRP3 inflammasome activation can abrogate the alleviated effect of LXRβ/ABCA1 pathway on the senescence of cochlear hair cells but did not affect the expression of LXRβ/ABCA1.Our study are the first to demonstrate that abnormal lipid droplet accumulation and decreased LXRβ/ABCA1 pathway are observed in cochlear hair cells following the occurrence of age-related hearing loss. Upregulation of LXRβ/ABCA1 in senescent cochlear hair cells can reduce lipid droplet accumulation in cochlear hair cells and alleviate their senescence, which may be related to the inhibition of NLRP3 inflammasome activation. These findings provide potential targets for the treatment of age-related hearing loss.
Collapse
Affiliation(s)
- Dongye Guo
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jichang Wu
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenling Shen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andi Zhang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyuan Zou
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaili Chen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiyi Huang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Pan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilin Shen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peilin Ji
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Wen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Kong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bin Ye
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
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
|
19
|
Mishra S, Swain PS, Pati S, Dehury B. Extracellular domain of TREM2 possess two distinct ligand recognition sites: Insights from machine-learning guided docking and all-atoms molecular dynamics simulations. Heliyon 2025; 11:e41414. [PMID: 39866401 PMCID: PMC11759634 DOI: 10.1016/j.heliyon.2024.e41414] [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: 06/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
The myeloid-specific triggering receptors expressed on myeloid cells 2 (TREM2) is a group of class I receptors expressed in brain microglia plays a decisive role in neurodegenerative diseases such as Alzheimer's disease (AD) and Nasu Hakola disease (NHD). The extracellular domain (ECD) of TREM2 interacts with a wide-range of ligands, yet the molecular mechanism underlying recognition of such ligands to this class I receptor remains underexplored. Herein, we undertook a systematic investigation for exploring the mode of ligand recognition in immunoglobulin-like ectodomain by employing both knowledge-based and machine-learning guided molecular docking approach followed by the state-of-the-art all atoms molecular dynamics (MD) simulations. Besides the known binding site formed by complementarity-determining regions (CDR) 1 and CDR2 loops, which enables the binding of different anionic ligands, our study identifies the presence of second binding site formed by β-strands towards the C-terminal end. We observe a dense network of hydrophobic contacts formed between the explored ligands and CDR loops and β-strands, specifically CDR1, CDR2, β-strand C', loop connecting β-strand D and E, and loop connecting β-strand E and F. Ligand binding in immunoglobulin-like ectodomain increases the conformational flexibility of CDR2 loop, thus most frequently observed pathogenic variants i.e. R47H and R62H in TREM2 may affect the development and progression of AD. Our knowledge-based and machine-learning guided docking and physics-based simulations study unveils deep insights into the endogenous ligand recognition by the positive surface ligand binding site and distant core site pave the way for exploration of other small molecules towards development of novel therapeutics against Alzheimer's disease.
Collapse
Affiliation(s)
- Sarbani Mishra
- ICMR-regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India
| | - Preety Sthutika Swain
- ICMR-regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India
| | - Sanghamitra Pati
- ICMR-regional Medical Research Centre, Nalco Square, Chandrasekharpur, Bhubaneswar, 751023, Odisha, India
| | - Budheswar Dehury
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
20
|
Gharibani P, Abramson E, Shanmukha S, Smith MD, Godfrey WH, Lee JJ, Hu J, Baydyuk M, Dorion MF, Deng X, Guo Y, Calle AJ, A Hwang S, Huang JK, Calabresi PA, Kornberg MD, Kim PM. The protein kinase C modulator bryostatin-1 therapeutically targets microglia to attenuate neuroinflammation and promote remyelination. Sci Transl Med 2025; 17:eadk3434. [PMID: 39772770 DOI: 10.1126/scitranslmed.adk3434] [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: 08/18/2023] [Revised: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In multiple sclerosis (MS), microglia and macrophages within the central nervous system (CNS) play an important role in determining the balance among demyelination, neurodegeneration, and myelin repair. Phagocytic and regenerative functions of these CNS innate immune cells support remyelination, whereas chronic and maladaptive inflammatory activation promotes lesion expansion and disability, particularly in the progressive forms of MS. No currently approved drugs convincingly target microglia and macrophages within the CNS, contributing to the lack of therapies aimed at promoting remyelination and slowing disease progression for individuals with MS. Here, we found that the protein kinase C (PKC)-modulating drug bryostatin-1 (bryo-1), a CNS-penetrant compound with an established human safety profile, shifts the transcriptional programs of microglia and CNS-associated macrophages from a proinflammatory phenotype to a regenerative phenotype in vitro and in vivo. Treatment of microglia with bryo-1 stimulated scavenger pathways, phagocytosis, and secretion of factors that prevented the activation of neuroinflammatory reactive astrocytes while also promoting neuroaxonal health and oligodendrocyte differentiation. In line with these findings, systemic treatment of mice with bryo-1 augmented remyelination after a focal demyelinating injury. Our results demonstrate the potential of bryo-1 and possibly a wider class of PKC modulators as myelin-regenerative and supportive agents in MS and other neurologic diseases.
Collapse
Affiliation(s)
- Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Efrat Abramson
- Interdepartmental Neuroscience Program, Yale University School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wesley H Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Judy J Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jingwen Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew J Calle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Soonmyung A Hwang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
21
|
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
|
22
|
Huynh TN, Fikse EN, De La Torre AL, Havrda MC, Chang CCY, Chang TY. Inhibiting the Cholesterol Storage Enzyme ACAT1/SOAT1 in Aging Apolipoprotein E4 Mice Alters Their Brains' Inflammatory Profiles. Int J Mol Sci 2024; 25:13690. [PMID: 39769453 PMCID: PMC11727783 DOI: 10.3390/ijms252413690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Aging and apolipoprotein E4 (APOE4) are the two most significant risk factors for late-onset Alzheimer's disease (LOAD). Compared to APOE3, APOE4 disrupts cholesterol homeostasis, increases cholesteryl esters (CEs), and exacerbates neuroinflammation in brain cells, including microglia. Targeting CEs and neuroinflammation could be a novel strategy to ameliorate APOE4-dependent phenotypes. Toll-like receptor 4 (TLR4) is a key macromolecule in inflammation, and its regulation is associated with the cholesterol content of lipid rafts in cell membranes. We previously demonstrated that in normal microglia expressing APOE3, inhibiting the cholesterol storage enzyme acyl-CoA:cholesterol acyltransferase 1 (ACAT1/SOAT1) reduces CEs, dampened neuroinflammation via modulating the fate of TLR4. We also showed that treating myelin debris-loaded normal microglia with ACAT inhibitor F12511 reduced cellular CEs and activated ABC transporter 1 (ABCA1) for cholesterol efflux. This study found that treating primary microglia expressing APOE4 with F12511 also reduces CEs, activates ABCA1, and dampens LPS-dependent NFκB activation. In vivo, two-week injections of nanoparticle F12511, which consists of DSPE-PEG2000, phosphatidylcholine, and F12511, to aged female APOE4 mice reduced TLR4 protein content and decreased proinflammatory cytokines, including IL-1β in mice brains. Overall, our work suggests nanoparticle F12511 is a novel agent to ameliorate LOAD.
Collapse
Affiliation(s)
- Thao N. Huynh
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA; (T.N.H.)
| | - Emma N. Fikse
- Department of Molecular and System Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Adrianna L. De La Torre
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA; (T.N.H.)
| | - Matthew C. Havrda
- Department of Molecular and System Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA; (T.N.H.)
| | - Ta Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA; (T.N.H.)
| |
Collapse
|
23
|
Zhu H, Hu E, Guo X, Yuan Z, Jiang H, Zhang W, Tang T, Wang Y, Li T. Promoting remyelination in central nervous system diseases: Potentials and prospects of natural products and herbal medicine. Pharmacol Res 2024; 210:107533. [PMID: 39617281 DOI: 10.1016/j.phrs.2024.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Myelin damage is frequently associated with central nervous system (CNS) diseases and is a critical factor influencing neurological function and disease prognosis. Nevertheless, the majority of current treatments for the CNS concentrate on gray matter injury and repair strategies, while clinical interventions specifically targeting myelin repair remain unavailable. In recent years, natural products and herbal medicine have achieved considerable progress in the domain of myelin repair, given their remarkable curative effect and low toxic side effects, demonstrating significant therapeutic potential. In this review, we present a rather comprehensive account of the mechanisms underlying myelin formation, injury, and repair, with a particular emphasis on the interactions between oligodendrocytes and other glial cells. Furthermore, we summarize the natural products and herbal medicine currently employed in remyelination along with their mechanisms of action, highlighting the potential and challenges of certain natural compounds to enhance myelin repair. This review aims to facilitate the expedited development of innovative therapeutics derived from natural products and herbal medicine and furnish novel insights into myelin repair in the CNS.
Collapse
Affiliation(s)
- Haonan Zhu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - En Hu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Xin Guo
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhiqiang Yuan
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Haoying Jiang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Tao Tang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Yang Wang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Teng Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China.
| |
Collapse
|
24
|
Jian H, Wu K, Lv Y, Du J, Hou M, Zhang C, Gao J, Zhou H, Feng S. A critical role for microglia in regulating metabolic homeostasis and neural repair after spinal cord injury. Free Radic Biol Med 2024; 225:469-481. [PMID: 39413980 DOI: 10.1016/j.freeradbiomed.2024.10.288] [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: 09/13/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Traumatic spinal cord injury (SCI) often results in severe immune and metabolic disorders, aggravating neurological damage and inhibiting locomotor functional recovery. Microglia, as resident immune cells of the spinal cord, play crucial roles in maintaining neural homeostasis under physiological conditions. However, the precise role of microglia in regulating immune and metabolic functions in SCI is still unclear and is easily confused with that of macrophages. In this study, we pharmacologically depleted microglia to explore the role of microglia after SCI. We found that microglia are beneficial for the recovery of locomotor function. Depleting microglia disrupted glial scar formation, reducing neurogenesis and angiogenesis. Using liquid chromatography tandem mass spectrometry (LC‒MS/MS), we discovered that depleting microglia significantly inhibits lipid metabolism processes such as fatty acid degradation, unsaturated fatty acid biosynthesis, glycophospholipid metabolism, and sphingolipid metabolism, accompanied by the accumulation of multiple organic acids. Subsequent studies demonstrated that microglial depletion increased the inhibition of FASN after SCI. FASN inhibition exacerbated malonyl-CoA accumulation and significantly impeded the activity of mTORC1. Moreover, microglial depletion exacerbated the oxidative stress of neurons. In summary, our results indicate that microglia alleviate neural damage and metabolic disorders after SCI, which is beneficial for achieving optimal neuroprotection and neural repair.
Collapse
Affiliation(s)
- Huan Jian
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Kailin Wu
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Yigang Lv
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Jiawei Du
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Mengfan Hou
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Chi Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Hengxing Zhou
- Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Orthopedics, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
25
|
Wu J, Kislinger G, Duschek J, Durmaz AD, Wefers B, Feng R, Nalbach K, Wurst W, Behrends C, Schifferer M, Simons M. Nonvesicular lipid transfer drives myelin growth in the central nervous system. Nat Commun 2024; 15:9756. [PMID: 39528474 PMCID: PMC11554831 DOI: 10.1038/s41467-024-53511-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Oligodendrocytes extend numerous cellular processes that wrap multiple times around axons to generate lipid-rich myelin sheaths. Myelin biogenesis requires an enormously productive biosynthetic machinery for generating and delivering these large amounts of newly synthesized lipids. Yet, a complete understanding of this process remains elusive. Utilizing volume electron microscopy, we demonstrate that the oligodendroglial endoplasmic reticulum (ER) is enriched in developing myelin, extending into and making contact with the innermost myelin layer where growth occurs. We explore the possibility of transfer of lipids from the ER to myelin, and find that the glycolipid transfer protein (GLTP), implicated in nonvesicular lipid transport, is highly enriched in the growing myelin sheath. Mice with a specific knockout of Gltp in oligodendrocytes exhibit ER pathology, hypomyelination and a decrease in myelin glycolipid content. In summary, our results demonstrate a role for nonvesicular lipid transport in CNS myelin growth, revealing a cellular pathway in developmental myelination.
Collapse
Affiliation(s)
- Jianping Wu
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Georg Kislinger
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Jerome Duschek
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Ayşe Damla Durmaz
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases, Munich, Germany
| | - Ruoqing Feng
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Graduate School of Systemic Neurosciences, LMU Munich, Munich, Germany
| | - Karsten Nalbach
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg, Germany
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Christian Behrends
- Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
26
|
Gao R, Song SJ, Tian MY, Wang LB, Zhang Y, Li X. Myelin debris phagocytosis in demyelinating disease. Glia 2024; 72:1934-1954. [PMID: 39073200 DOI: 10.1002/glia.24602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Demyelinating diseases are often caused by a variety of triggers, including immune responses, viral infections, malnutrition, hypoxia, or genetic factors, all of which result in the loss of myelin in the nervous system. The accumulation of myelin debris at the lesion site leads to neuroinflammation and inhibits remyelination; therefore, it is crucial to promptly remove the myelin debris. Initially, Fc and complement receptors on cellular surfaces were the primary clearance receptors responsible for removing myelin debris. However, subsequent studies have unveiled the involvement of additional receptors, including Mac-2, TAM receptors, and the low-density lipoprotein receptor-related protein 1, in facilitating the removal process. In addition to microglia and macrophages, which serve as the primary effector cells in the disease phase, a variety of other cell types such as astrocytes, Schwann cells, and vascular endothelial cells have been demonstrated to engage in the phagocytosis of myelin debris. Furthermore, we have concluded that oligodendrocyte precursor cells, as myelination precursor cells, also exhibit this phagocytic capability. Moreover, our research group has innovatively identified the low-density lipoprotein receptor as a potential phagocytic receptor for myelin debris. In this article, we discuss the functional processes of various phagocytes in demyelinating diseases. We also highlight the alterations in signaling pathways triggered by phagocytosis, and provide a comprehensive overview of the various phagocytic receptors involved. Such insights are invaluable for pinpointing potential therapeutic strategies for the treatment of demyelinating diseases by targeting phagocytosis.
Collapse
Affiliation(s)
- Rui Gao
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Sheng-Jiao Song
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Meng-Yuan Tian
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Li-Bin Wang
- Neurosurgery Department, Huazhong University of Science and Technology Union Shenzhen Hospital/Shenzhen Nanshan Hospital, Shenzhen, Guangdong, China
| | - Yuan Zhang
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xing Li
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
27
|
Huynh TN, Fikse EN, Havrda MC, Chang CCY, Chang TY. Inhibiting the cholesterol storage enzyme ACAT1/SOAT1 in aging Apolipoprotein E4 mice alter their brains inflammatory profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620063. [PMID: 39484620 PMCID: PMC11527143 DOI: 10.1101/2024.10.24.620063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Aging and Apolipoprotein E4 (APOE4) are the two most significant risk factors for late-onset Alzheimer's disease (LOAD). Compared to APOE3, APOE4 disrupts cholesterol homeostasis, increases cholesteryl esters (CEs), and exacerbates neuroinflammation in brain cells including microglia. Targeting CEs and neuroinflammation could be a novel strategy to ameliorate APOE4 dependent phenotypes. Toll-like receptor 4 (TLR4) is a key player in inflammation, its regulation is associated with cholesterol content of lipid rafts in cell membranes. We previously demonstrated that in normal microglia expressing APOE3, inhibiting the cholesterol storage enzyme acylCoA:cholesterol acyltransferase 1 (ACAT1/SOAT1) reduces CEs, dampened neuroinflammation via modulating the fate of TLR4. We also showed that treating myelin debris-loaded normal microglia with ACAT inhibitor F12511 reduced cellular CEs and activated ABC transporter 1 (ABCA1) for cholesterol efflux. In this study, we found that treating primary microglia expressing APOE4 with F12511 also reduces CEs, activated ABCA1, and dampened LPS dependent NFkB activation. In vivo, a two-week injections of nanoparticle F12511, which consists of DSPE-PEG 2000 , phosphatidylcholine, and F12511, to aged female APOE4 mice reduced TLR4 protein content and decreased proinflammatory cytokines including IL-1β in APOE4 mice brains. Overall, our work suggests nanoparticle F12511 is a novel agent to ameliorate LOAD.
Collapse
|
28
|
Huynh TN, Havrda MC, Zanazzi GJ, Chang CCY, Chang TY. Inhibiting the Cholesterol Storage Enzyme ACAT1/SOAT1 in Myelin Debris-Treated Microglial Cell Lines Activates the Gene Expression of Cholesterol Efflux Transporter ABCA1. Biomolecules 2024; 14:1301. [PMID: 39456234 PMCID: PMC11505751 DOI: 10.3390/biom14101301] [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: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Aging is the major risk factor for Alzheimer's disease (AD). In the aged brain, myelin debris accumulates and is cleared by microglia. Phagocytosed myelin debris increases neutral lipid droplet content in microglia. Neutral lipids include cholesteryl esters (CE) and triacylglycerol (TAG). To examine the effects of myelin debris on neutral lipid content in microglia, we added myelin debris to human HMC3 and mouse N9 cells. The results obtained when using 3H-oleate as a precursor in intact cells reveal that myelin debris significantly increases the biosynthesis of CE but not TAG. Mass analyses have shown that myelin debris increases both CE and TAG. The increase in CE biosynthesis was abolished using inhibitors of the cholesterol storage enzyme acyl-CoA:cholesterol acyltransferase 1 (ACAT1/SOAT1). ACAT1 inhibitors are promising drug candidates for AD treatment. In myelin debris-loaded microglia, treatment with two different ACAT1 inhibitors, K604 and F12511, increased the mRNA and protein content of ATP-binding cassette subfamily A1 (ABCA1), a protein that is located at the plasma membrane and which controls cellular cholesterol disposal. The effect of the ACAT1 inhibitor on ABCA1 was abolished by preincubating cells with the liver X receptor (LXR) antagonist GSK2033. We conclude that ACAT1 inhibitors prevent the accumulation of cholesterol and CE in myelin debris-treated microglia by activating ABCA1 gene expression via the LXR pathway.
Collapse
Affiliation(s)
- Thao N. Huynh
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - Matthew C. Havrda
- Department of Molecular and System Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - George J. Zanazzi
- Department of Pathology and Laboratory Medicine, Dartmouth–Hitchcock Medical Center, Lebanon, NH 03766, USA;
| | - Catherine C. Y. Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| | - Ta Yuan Chang
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA;
| |
Collapse
|
29
|
Gogoleva VS, Mundt S, De Feo D, Becher B. Mononuclear phagocytes in autoimmune neuroinflammation. Trends Immunol 2024; 45:814-823. [PMID: 39307582 DOI: 10.1016/j.it.2024.08.005] [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: 07/08/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 10/13/2024]
Abstract
A healthy mammalian central nervous system (CNS) harbors a diverse population of leukocytes including members of the mononuclear phagocyte system (MPS). Exerting their specific functions, CNS tissue-resident macrophages as well as associated monocytes and dendritic cells (DCs) maintain CNS homeostasis. Under neuroinflammatory conditions, leukocytes from the systemic immune compartment invade the CNS. This review focuses on the newly discovered roles of the MPS in autoimmune neuroinflammation elicited by encephalitogenic T cells. We propose that CNS-associated DCs act as gatekeepers and antigen-presenting cells that guide the adaptive immune response while bone marrow (BM)-derived monocytes contribute to immunopathology and tissue damage. By contrast, CNS-resident macrophages primarily support tissue function and promote the repair and maintenance of CNS functions.
Collapse
Affiliation(s)
- Violetta S Gogoleva
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Hoffmann A, Miron VE. CNS macrophage contributions to myelin health. Immunol Rev 2024; 327:53-70. [PMID: 39484853 DOI: 10.1111/imr.13416] [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] [Indexed: 11/03/2024]
Abstract
Myelin is the membrane surrounding neuronal axons in the central nervous system (CNS), produced by oligodendrocytes to provide insulation for electrical impulse conduction and trophic/metabolic support. CNS dysfunction occurs following poor development of myelin in infancy, myelin damage in neurological diseases, and impaired regeneration of myelin with disease progression in aging. The lack of approved therapies aimed at supporting myelin health highlights the critical need to identify the cellular and molecular influences on oligodendrocytes. CNS macrophages have been shown to influence the development, maintenance, damage and regeneration of myelin, revealing critical interactions with oligodendrocyte lineage cells. CNS macrophages are comprised of distinct populations, including CNS-resident microglia and cells associated with CNS border regions (the meninges, vasculature, and choroid plexus), in addition to macrophages derived from monocytes infiltrating from the blood. Importantly, the distinct contribution of these macrophage populations to oligodendrocyte lineage responses and myelin health are only just beginning to be uncovered, with the advent of new tools to specifically identify, track, and target macrophage subsets. Here, we summarize the current state of knowledge on the roles of CNS macrophages in myelin health, and recent developments in distinguishing the roles of macrophage populations in development, homeostasis, and disease.
Collapse
Affiliation(s)
- Alana Hoffmann
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
31
|
Hanlon MM, Smith CM, Canavan M, Neto NGB, Song Q, Lewis MJ, O’Rourke AM, Tynan O, Barker BE, Gallagher P, Mullan R, Hurson C, Moran B, Monaghan MG, Pitzalis C, Fletcher JM, Nagpal S, Veale DJ, Fearon U. Loss of synovial tissue macrophage homeostasis precedes rheumatoid arthritis clinical onset. SCIENCE ADVANCES 2024; 10:eadj1252. [PMID: 39321281 PMCID: PMC11423874 DOI: 10.1126/sciadv.adj1252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
This study performed an in-depth investigation into the myeloid cellular landscape in the synovium of patients with rheumatoid arthritis (RA), "individuals at risk" of RA, and healthy controls (HC). Flow cytometric analysis demonstrated the presence of a CD40-expressing CD206+CD163+ macrophage population dominating the inflamed RA synovium, associated with disease activity and treatment response. In-depth RNA sequencing and metabolic analysis demonstrated that this macrophage population is transcriptionally distinct, displaying unique inflammatory and tissue-resident gene signatures, has a stable bioenergetic profile, and regulates stromal cell responses. Single-cell RNA sequencing profiling of 67,908 RA and HC synovial tissue cells identified nine transcriptionally distinct macrophage clusters. IL-1B+CCL20+ and SPP1+MT2A+ are the principal macrophage clusters in RA synovium, displaying heightened CD40 gene expression, capable of shaping stromal cell responses, and are importantly enriched before disease onset. Combined, these findings identify the presence of an early pathogenic myeloid signature that shapes the RA joint microenvironment and represents a unique opportunity for early diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Megan M. Hanlon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Conor M. Smith
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
- Translational Immunopathology, School of Biochemistry and Immunology and School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nuno G. B. Neto
- Department of Mechanical and Manufacturing Engineering, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Qingxuan Song
- Immunology and Discovery Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC and Barts Health NHS Trust, London, UK
| | - Aoife M. O’Rourke
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
- Translational Immunopathology, School of Biochemistry and Immunology and School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Orla Tynan
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Brianne E. Barker
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Phil Gallagher
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ronan Mullan
- Department of Rheumatology, Adelaide and Meath Hospital, Dublin, Ireland
| | - Conor Hurson
- Department of Orthopaedics, St. Vincent’s University Hospital, Dublin, Ireland
| | - Barry Moran
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Michael G. Monaghan
- Department of Mechanical and Manufacturing Engineering, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London and Barts NIHR BRC and Barts Health NHS Trust, London, UK
- Department of Biomedical Sciences, Humanitas University and Humanitas Research Hospital, Milan, Italy
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sunil Nagpal
- Immunology and Discovery Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Douglas J. Veale
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
32
|
McCray TJ, Bedford LM, Bissel SJ, Lamb BT. Trem2-deficiency aggravates and accelerates age-related myelin degeneration. Acta Neuropathol Commun 2024; 12:154. [PMID: 39300502 PMCID: PMC11411802 DOI: 10.1186/s40478-024-01855-3] [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: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/22/2024] Open
Abstract
Aging is the greatest known risk factor for most neurodegenerative diseases. Myelin degeneration is an early pathological indicator of these diseases and a normal part of aging; albeit, to a lesser extent. Despite this, little is known about the contribution of age-related myelin degeneration on neurodegenerative disease. Microglia participate in modulating white matter events from demyelination to remyelination, including regulation of (de)myelination by the microglial innate immune receptor triggering receptor expressed on myeloid cells 2 (TREM2). Here, we demonstrate Trem2-deficiency aggravates and accelerates age-related myelin degeneration in the striatum. We show TREM2 is necessary for remyelination by recruiting reparative glia and mediating signaling that promotes OPC differentiation/maturation. In response to demyelination, TREM2 is required for phagocytosis of large volumes of myelin debris. In addition to lysosomal regulation, we show TREM2 can modify the ER stress response, even prior to overt myelin debris, that prevents lipid accumulation and microglial dysfunction. These data support a role for Trem2-dependent interactions in age-related myelin degeneration and suggest a basis for how early dysfunctional microglia could contribute to disease pathology through insufficent repair, defective phagocytosis, and the ER stress response.
Collapse
Affiliation(s)
- Tyler J McCray
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Logan M Bedford
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Stephanie J Bissel
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Bruce T Lamb
- Stark Neurosciences Research Institute, Indiana University, School of Medicine, Indianapolis, IN 46202, USA.
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
33
|
Cao Y, Zhao LW, Chen ZX, Li SH. New insights in lipid metabolism: potential therapeutic targets for the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1430465. [PMID: 39323915 PMCID: PMC11422391 DOI: 10.3389/fnins.2024.1430465] [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: 05/10/2024] [Accepted: 08/14/2024] [Indexed: 09/27/2024] Open
Abstract
Alzheimer's disease (AD) is increasingly recognized as being intertwined with the dysregulation of lipid metabolism. Lipids are a significant class of nutrients vital to all organisms, playing crucial roles in cellular structure, energy storage, and signaling. Alterations in the levels of various lipids in AD brains and dysregulation of lipid pathways and transportation have been implicated in AD pathogenesis. Clinically, evidence for a high-fat diet firmly links disrupted lipid metabolism to the pathogenesis and progression of AD, although contradictory findings warrant further exploration. In view of the significance of various lipids in brain physiology, the discovery of complex and diverse mechanisms that connect lipid metabolism with AD-related pathophysiology will bring new hope for patients with AD, underscoring the importance of lipid metabolism in AD pathophysiology, and promising targets for therapeutic intervention. Specifically, cholesterol, sphingolipids, and fatty acids have been shown to influence amyloid-beta (Aβ) accumulation and tau hyperphosphorylation, which are hallmarks of AD pathology. Recent studies have highlighted the potential therapeutic targets within lipid metabolism, such as enhancing apolipoprotein E lipidation, activating liver X receptors and retinoid X receptors, and modulating peroxisome proliferator-activated receptors. Ongoing clinical trials are investigating the efficacy of these strategies, including the use of ketogenic diets, statin therapy, and novel compounds like NE3107. The implications of these findings suggest that targeting lipid metabolism could offer new avenues for the treatment and management of AD. By concentrating on alterations in lipid metabolism within the central nervous system and their contribution to AD development, this review aims to shed light on novel research directions and treatment approaches for combating AD, offering hope for the development of more effective management strategies.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lin-Wei Zhao
- Department of Cardiology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou University Central China Fuwai Hospital, Zhengzhou, China
| | - Zi-Xin Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shao-Hua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
34
|
Zota I, Chanoumidou K, Gravanis A, Charalampopoulos I. Stimulating myelin restoration with BDNF: a promising therapeutic approach for Alzheimer's disease. Front Cell Neurosci 2024; 18:1422130. [PMID: 39285941 PMCID: PMC11402763 DOI: 10.3389/fncel.2024.1422130] [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: 04/23/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Alzheimer's Disease (AD) is a chronic neurodegenerative disorder constituting the most common form of dementia (60%-70% of cases). Although AD presents majorly a neurodegenerative pathology, recent clinical evidence highlights myelin impairment as a key factor in disease pathogenesis. The lack of preventive or restorative treatment is emphasizing the need to develop novel therapeutic approaches targeting to the causes of the disease. Recent studies in animals and patients have highlighted the loss of myelination of the neuronal axons as an extremely aggravating factor in AD, in addition to the formation of amyloid plaques and neurofibrillary tangles that are to date the main pathological hallmarks of the disease. Myelin breakdown represents an early stage event in AD. However, it is still unclear whether myelin loss is attributed only to exogenous factors like inflammatory processes of the tissue or to impaired oligodendrogenesis as well. Neurotrophic factors are well established protective molecules under many pathological conditions of the neural tissue, contributing also to proper myelination. Due to their inability to be used as drugs, many research efforts are focused on substituting neurotrophic activity with small molecules. Our research team has recently developed novel micromolecular synthetic neurotrophin mimetics (MNTs), selectively acting on neurotrophin receptors, and thus offering a unique opportunity for innovative therapies against neurodegenerative diseases. These small sized, lipophilic molecules address the underlying biological effect of these diseases (neuroprotective action), but also they exert significant neurogenic actions inducing neuronal replacement of the disease areas. One of the significant neurotrophin molecules in the Central Nervous System is Brain-Derived-Neurotrophin-Factor (BDNF). BDNF is a neurotrophin that not only supports neuroprotection and adult neurogenesis, but also mediates pro-myelinating effects in the CNS. BDNF binds with high-affinity on the TrkB neurotrophin receptor and enhances myelination by increasing the density of oligodendrocyte progenitor cells (OPCs) and playing an important role in CNS myelination. Conclusively, in the present review, we discuss the myelin pathophysiology in Alzheimer's Diseases, as well as the role of neurotrophins, and specifically BDNF, in myelin maintenance and restoration, revealing its valuable therapeutic potential against AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas (IMBB-FORTH), Heraklion, Greece
| |
Collapse
|
35
|
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
|
36
|
Raffaele S, Nguyen N, Milanese M, Mannella FC, Boccazzi M, Frumento G, Bonanno G, Abbracchio MP, Bonifacino T, Fumagalli M. Montelukast improves disease outcome in SOD1 G93A female mice by counteracting oligodendrocyte dysfunction and aberrant glial reactivity. Br J Pharmacol 2024; 181:3303-3326. [PMID: 38751168 DOI: 10.1111/bph.16408] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/08/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive motor neuron (MN) loss and consequent muscle atrophy, for which no effective therapies are available. Recent findings reveal that disease progression is fuelled by early aberrant neuroinflammation and the loss of oligodendrocytes with neuroprotective and remyelinating properties. On this basis, pharmacological interventions capable of restoring a pro-regenerative local milieu and re-establish proper oligodendrocyte functions may be beneficial. EXPERIMENTAL APPROACH Here, we evaluated the in vivo therapeutic effects of montelukast (MTK), an antagonist of the oligodendroglial G protein-coupled receptor 17 (GPR17) and of cysteinyl-leukotriene receptor 1 (CysLT1R) receptors on microglia and astrocytes, in the SOD1G93A ALS mouse model. We chronically treated SOD1G93A mice with MTK, starting from the early symptomatic disease stage. Disease progression was assessed by behavioural and immunohistochemical approaches. KEY RESULTS Oral MTK treatment significantly extended survival probability, delayed body weight loss and ameliorated motor functionalityonly in female SOD1G93A mice. Noteworthy, MTK significantly restored oligodendrocyte maturation and induced significant changes in the reactive phenotype and morphological features of microglia/macrophages and astrocytes in the spinal cord of female SOD1G93A mice, suggesting enhanced pro-regenerative functions. Importantly, concomitant MN preservation has been detected after MTK administration. No beneficial effects were observed in male mice, highlighting a sex-based difference in the protective activity of MTK. CONCLUSIONS AND IMPLICATIONS Our results provide the first preclinical evidence indicating that repurposing of MTK, a safe and marketed anti-asthmatic drug, may be a promising sex-specific strategy for personalized ALS treatment.
Collapse
Affiliation(s)
- Stefano Raffaele
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Nhung Nguyen
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca C Mannella
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Giulia Frumento
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, Università degli Studi di Genova, Genoa, Italy
- Inter-University Center for the Promotion of the 3R Principles in Teaching and Research (Centro 3R), Pisa, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Deng Q, Wu C, Parker E, Liu TCY, Duan R, Yang L. Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances. Aging Dis 2024; 15:1537-1564. [PMID: 37815901 PMCID: PMC11272214 DOI: 10.14336/ad.2023.0907] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Alzheimer's disease, one of the most common forms of dementia, is characterized by a slow progression of cognitive impairment and neuronal loss. Currently, approved treatments for AD are hindered by various side effects and limited efficacy. Despite considerable research, practical treatments for AD have not been developed. Increasing evidence shows that glial cells, especially microglia and astrocytes, are essential in the initiation and progression of AD. During AD progression, activated resident microglia increases the ability of resting astrocytes to transform into reactive astrocytes, promoting neurodegeneration. Extensive clinical and molecular studies show the involvement of microglia and astrocyte-mediated neuroinflammation in AD pathology, indicating that microglia and astrocytes may be potential therapeutic targets for AD. This review will summarize the significant and recent advances of microglia and astrocytes in the pathogenesis of AD in three parts. First, we will review the typical pathological changes of AD and discuss microglia and astrocytes in terms of function and phenotypic changes. Second, we will describe microglia and astrocytes' physiological and pathological role in AD. These roles include the inflammatory response, "eat me" and "don't eat me" signals, Aβ seeding, propagation, clearance, synapse loss, synaptic pruning, remyelination, and demyelination. Last, we will review the pharmacological and non-pharmacological therapies targeting microglia and astrocytes in AD. We conclude that microglia and astrocytes are essential in the initiation and development of AD. Therefore, understanding the new role of microglia and astrocytes in AD progression is critical for future AD studies and clinical trials. Moreover, pharmacological, and non-pharmacological therapies targeting microglia and astrocytes, with specific studies investigating microglia and astrocyte-mediated neuronal damage and repair, may be a promising research direction for future studies regarding AD treatment and prevention.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
38
|
Etxeberria A, Shen YAA, Vito S, Silverman SM, Imperio J, Lalehzadeh G, Soung AL, Du C, Xie L, Choy MK, Hsiao YC, Ngu H, Cho CH, Ghosh S, Novikova G, Rezzonico MG, Leahey R, Weber M, Gogineni A, Elstrott J, Xiong M, Greene JJ, Stark KL, Chan P, Roth GA, Adrian M, Li Q, Choi M, Wong WR, Sandoval W, Foreman O, Nugent AA, Friedman BA, Sadekar S, Hötzel I, Hansen DV, Chih B, Yuen TJ, Weimer RM, Easton A, Meilandt WJ, Bohlen CJ. Neutral or Detrimental Effects of TREM2 Agonist Antibodies in Preclinical Models of Alzheimer's Disease and Multiple Sclerosis. J Neurosci 2024; 44:e2347232024. [PMID: 38830764 PMCID: PMC11255434 DOI: 10.1523/jneurosci.2347-23.2024] [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: 12/18/2023] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/05/2024] Open
Abstract
Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-β (Aβ) pathology (PS2APP) or combined Aβ and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.
Collapse
Affiliation(s)
- Ainhoa Etxeberria
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Yun-An A Shen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Stephen Vito
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Sean M Silverman
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jose Imperio
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Guita Lalehzadeh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Allison L Soung
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Changchun Du
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Luke Xie
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Man Kin Choy
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Yi-Chun Hsiao
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Hai Ngu
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Chang Hoon Cho
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Soumitra Ghosh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Gloriia Novikova
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | | | - Rebecca Leahey
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Martin Weber
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Alvin Gogineni
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Justin Elstrott
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Monica Xiong
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jacob J Greene
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Kimberly L Stark
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Pamela Chan
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Gillie A Roth
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Qingling Li
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Meena Choi
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Weng Ruh Wong
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Wendy Sandoval
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Oded Foreman
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Alicia A Nugent
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Brad A Friedman
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Isidro Hötzel
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - David V Hansen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Ben Chih
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Tracy J Yuen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Robby M Weimer
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Amy Easton
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - William J Meilandt
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher J Bohlen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
39
|
Ou Z, Cheng Y, Ma H, Chen K, Lin Q, Chen J, Guo R, Huang Z, Cheng Q, Alaeiilkhchi N, Zhu Q, Huang Z, Jiang H. miR-223 accelerates lipid droplets clearance in microglia following spinal cord injury by upregulating ABCA1. J Transl Med 2024; 22:659. [PMID: 39010173 PMCID: PMC11247820 DOI: 10.1186/s12967-024-05480-5] [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: 01/30/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is characterized by extensive demyelination and inflammatory responses. Facilitating the clearance of lipid droplets (LDs) within microglia contributes to creating a microenvironment that favors neural recovery and provides essential materials for subsequent remyelination. Therefore, investigating MicroRNAs (miRNAs) that regulate lipid homeostasis after SCI and elucidating their potential mechanisms in promoting LDs clearance in microglia have become focal points of SCI research. METHODS We established a subacute C5 hemicontusion SCI model in mice and performed transcriptomic sequencing on the injury epicenter to identify differentially expressed genes and associated pathways. Confocal imaging was employed to observe LDs accumulation. Multi-omics analyses were conducted to identify differentially expressed mRNA and miRNA post-SCI. Pathway enrichment analysis and protein-protein interaction network construction were performed using bioinformatics methods, revealing miR-223-Abca1 as a crucial miRNA-mRNA pair in lipid metabolism regulation. BV2 microglia cell lines overexpressing miR-223 were engineered, and immunofluorescence staining, western blot, and other techniques were employed to assess LDs accumulation, relevant targets, and inflammatory factor expression, confirming its role in regulating lipid homeostasis in microglia. RESULTS Histopathological results of our hemicontusion SCI model confirmed LDs aggregation at the injury epicenter, predominantly within microglia. Our transcriptomic analysis during the subacute phase of SCI in mice implicated ATP-binding cassette transporter A1 (Abca1) as a pivotal gene in lipid homeostasis, cholesterol efflux and microglial activation. Integrative mRNA-miRNA multi-omics analysis highlighted the crucial role of miR-223 in the neuroinflammation process following SCI, potentially through the regulation of lipid metabolism via Abca1. In vitro experiments using BV2 cells overexpressing miR-223 demonstrated that elevated levels of miR-223 enhance ABCA1 expression in myelin debris and LPS-induced BV2 cells. This promotes myelin debris degradation and LDs clearance, and induces a shift toward an anti-inflammatory M2 phenotype. CONCLUSIONS In summary, our study unveils the critical regulatory role of miR-223 in lipid homeostasis following SCI. The mechanism by which this occurs involves the upregulation of ABCA1 expression, which facilitates LDs clearance and myelin debris degradation, consequently alleviating the lipid burden, and inhibiting inflammatory polarization of microglia. These findings suggest that strategies to enhance miR-223 expression and target ABCA1, thereby augmenting LDs clearance, may emerge as appealing new clinical targets for SCI treatment.
Collapse
Affiliation(s)
- Zhilin Ou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yongquan Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiong Lin
- School of Anesthesiology, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jiayu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ruqin Guo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiping Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qixian Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Nima Alaeiilkhchi
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, Vancouver, Canada
| | - Qingan Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zucheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
40
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [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: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
41
|
Zhou LQ, Chu YH, Dong MH, Yang S, Chen M, Tang Y, Pang XW, You YF, Wu LJ, Wang W, Qin C, Tian DS. Ldl-stimulated microglial activation exacerbates ischemic white matter damage. Brain Behav Immun 2024; 119:416-430. [PMID: 38636563 DOI: 10.1016/j.bbi.2024.04.014] [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: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
The role of microglia in triggering the blood-brain barrier (BBB) impairment and white matter damage after chronic cerebral hypoperfusion is unclear. Here we demonstrated that the vessel-adjacent microglia were specifically activated by the leakage of plasma low-density lipoprotein (LDL), which led to BBB breakdown and ischemic demyelination. Interestingly, we found that LDL stimulation enhanced microglial phagocytosis, causing excessive engulfment of myelin debris and resulting in an overwhelming lipid burden in microglia. Surprisingly, these lipid-laden microglia exhibited a suppressed profile of inflammatory response and compromised pro-regenerative properties. Microglia-specific knockdown of LDLR or systematic medication lowering circulating LDL-C showed protective effects against ischemic demyelination. Overall, our findings demonstrated that LDL-stimulated vessel-adjacent microglia possess a disease-specific molecular signature, characterized by suppressed regenerative properties, which is associated with the propagation of demyelination during ischemic white matter damage.
Collapse
Affiliation(s)
- Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
42
|
Mallick K, Paul S, Banerjee S, Banerjee S. Lipid Droplets and Neurodegeneration. Neuroscience 2024; 549:13-23. [PMID: 38718916 DOI: 10.1016/j.neuroscience.2024.04.014] [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: 03/01/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/15/2024]
Abstract
Energy metabolism in the brain has been considered one of the critical research areas of neuroscience for ages. One of the most vital parts of brain metabolism cascades is lipid metabolism, and fatty acid plays a crucial role in this process. The fatty acid breakdown process in mitochondria undergoes through a conserved pathway known as β-oxidation where acetyl-CoA and shorter fatty acid chains are produced along with a significant amount of energy molecule. Further, the complete breakdown of fatty acids occurs when they enter the mitochondrial oxidative phosphorylation. Cells store energy as neutral lipids in organelles known as Lipid Droplets (LDs) to prepare for variations in the availability of nutrients. Fatty acids are liberated by lipid droplets and are transported to various cellular compartments for membrane biogenesis or as an energy source. Current research shows that LDs are important in inflammation, metabolic illness, and cellular communication. Lipid droplet biology in peripheral organs like the liver and heart has been well investigated, while the brain's LDs have received less attention. Recently, there has been increased awareness of the existence and role of these dynamic organelles in the central nervous system, mainly connected to neurodegeneration. In this review, we discussed the role of beta-oxidation and lipid droplet formation in the oxidative phosphorylation process, which directly affects neurodegeneration through various pathways.
Collapse
Affiliation(s)
- Keya Mallick
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, India.
| | - Shuchismita Paul
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, India.
| | - Sayani Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, India.
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, India.
| |
Collapse
|
43
|
Beiter RM, Sheehan PW, Schafer DP. Microglia phagocytic mechanisms: Development informing disease. Curr Opin Neurobiol 2024; 86:102877. [PMID: 38631077 PMCID: PMC11162951 DOI: 10.1016/j.conb.2024.102877] [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: 12/15/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Microglia are tissue-resident macrophages and professional phagocytes of the central nervous system (CNS). In development, microglia-mediated phagocytosis is important for sculpting the cellular architecture. This includes the engulfment of dead/dying cells, pruning extranumerary synapses and axons, and phagocytosing fragments of myelin sheaths. Intriguingly, these developmental phagocytic mechanisms by which microglia sculpt the CNS are now appreciated as important for eliminating synapses, myelin, and proteins during neurodegeneration. Here, we discuss parallels between neurodevelopment and neurodegeneration, which highlights how development is informing disease. We further discuss recent advances and challenges towards therapeutically targeting these phagocytic pathways and how we can leverage development to overcome these challenges.
Collapse
Affiliation(s)
- Rebecca M Beiter
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Patrick W Sheehan
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
44
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
45
|
Muzio L, Perego J. CNS Resident Innate Immune Cells: Guardians of CNS Homeostasis. Int J Mol Sci 2024; 25:4865. [PMID: 38732082 PMCID: PMC11084235 DOI: 10.3390/ijms25094865] [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: 03/21/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Although the CNS has been considered for a long time an immune-privileged organ, it is now well known that both the parenchyma and non-parenchymal tissue (meninges, perivascular space, and choroid plexus) are richly populated in resident immune cells. The advent of more powerful tools for multiplex immunophenotyping, such as single-cell RNA sequencing technique and upscale multiparametric flow and mass spectrometry, helped in discriminating between resident and infiltrating cells and, above all, the different spectrum of phenotypes distinguishing border-associated macrophages. Here, we focus our attention on resident innate immune players and their primary role in both CNS homeostasis and pathological neuroinflammation and neurodegeneration, two key interconnected aspects of the immunopathology of multiple sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Lab, IRCCS San Raffaele Scientific Institute, Institute of Experimental Neurology, 20133 Milan, Italy;
| | | |
Collapse
|
46
|
McCallum S, Suresh KB, Islam T, Saustad AW, Shelest O, Patil A, Lee D, Kwon B, Yenokian I, Kawaguchi R, Beveridge CH, Manchandra P, Randolph CE, Meares GP, Dutta R, Plummer J, Knott SRV, Chopra G, Burda JE. Lesion-remote astrocytes govern microglia-mediated white matter repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585251. [PMID: 38558977 PMCID: PMC10979953 DOI: 10.1101/2024.03.15.585251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Spared regions of the damaged central nervous system undergo dynamic remodeling and exhibit a remarkable potential for therapeutic exploitation. Here, lesion-remote astrocytes (LRAs), which interact with viable neurons, glia and neural circuitry, undergo reactive transformations whose molecular and functional properties are poorly understood. Using multiple transcriptional profiling methods, we interrogated LRAs from spared regions of mouse spinal cord following traumatic spinal cord injury (SCI). We show that LRAs acquire a spectrum of molecularly distinct, neuroanatomically restricted reactivity states that evolve after SCI. We identify transcriptionally unique reactive LRAs in degenerating white matter that direct the specification and function of local microglia that clear lipid-rich myelin debris to promote tissue repair. Fueling this LRA functional adaptation is Ccn1 , which encodes for a secreted matricellular protein. Loss of astrocyte CCN1 leads to excessive, aberrant activation of local microglia with (i) abnormal molecular specification, (ii) dysfunctional myelin debris processing, and (iii) impaired lipid metabolism, culminating in blunted debris clearance and attenuated neurological recovery from SCI. Ccn1 -expressing white matter astrocytes are specifically induced by local myelin damage and generated in diverse demyelinating disorders in mouse and human, pointing to their fundamental, evolutionarily conserved role in white matter repair. Our findings show that LRAs assume regionally divergent reactivity states with functional adaptations that are induced by local context-specific triggers and influence disorder outcome. Astrocytes tile the central nervous system (CNS) where they serve vital roles that uphold healthy nervous system function, including regulation of synapse development, buffering of neurotransmitters and ions, and provision of metabolic substrates 1 . In response to diverse CNS insults, astrocytes exhibit disorder-context specific transformations that are collectively referred to as reactivity 2-5 . The characteristics of regionally and molecularly distinct reactivity states are incompletely understood. The mechanisms through which distinct reactivity states arise, how they evolve or resolve over time, and their consequences for local cell function and CNS disorder progression remain enigmatic. Immediately adjacent to CNS lesions, border-forming astrocytes (BFAs) undergo transcriptional reprogramming and proliferation to form a neuroprotective barrier that restricts inflammation and supports axon regeneration 6-9 . Beyond the lesion, spared but dynamic regions of the injured CNS exhibit varying degrees of synaptic circuit remodeling and progressive cellular responses to secondary damage that have profound consequences for neural repair and recovery 10,11 . Throughout these cytoarchitecturally intact, but injury-reactive regions, lesion-remote astrocytes (LRAs) intermingle with neurons and glia, undergo little to no proliferation, and exhibit varying degrees of cellular hypertrophy 7,12,13 . The molecular and functional properties of LRAs remain grossly undefined. Therapeutically harnessing spared regions of the injured CNS will require a clearer understanding of the accompanying cellular and molecular landscape. Here, we leveraged integrative transcriptional profiling methodologies to identify multiple spatiotemporally resolved, molecularly distinct states of LRA reactivity within the injured spinal cord. Computational modeling of LRA-mediated heterotypic cell interactions, astrocyte-specific conditional gene deletion, and multiple mouse models of acute and chronic CNS white matter degeneration were used to interrogate a newly identified white matter degeneration-reactive astrocyte subtype. We define how this reactivity state is induced and its role in governing the molecular and functional specification of local microglia that clear myelin debris from the degenerating white matter to promote repair.
Collapse
|
47
|
Li W, Meng X, Peng K, Han Y, Liu H, Zhao W, Wang G, Deng L, Liu H, Li Z, Ji F. Boosting Microglial Lipid Metabolism via TREM2 Signaling by Biomimetic Nanoparticles to Attenuate the Sevoflurane-Induced Developmental Neurotoxicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305989. [PMID: 38145349 PMCID: PMC10933683 DOI: 10.1002/advs.202305989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Lipid metabolism has been considered as a potential therapeutic target in sevoflurane-induced neurotoxicity that can potentially affect the learning and memory function in the developmental brain. Recently, triggering receptor expressed on myeloid cells 2 (TREM2) is identified as a crucial step in regulating lipid metabolism and associated with the pathogenesis of neurodegenerative diseases. Herein, it is reported that quercetin modified Cu2- x Se (abbreviated as CSPQ) nanoparticles can ameliorate sevoflurane-induced neurotoxicity by tuning the microglial lipid metabolism and promoting microglial M2-like polarization via TREM2 signaling pathway, in which the apolipoprotein E (ApoE), and adenosine triphosphate-binding cassette transporters (ABCA1 and ABCG1) levels are upregulated. Furthermore, the protective effects of CSPQ nanoparticles against sevoflurane-induced neurotoxicity via TREM2 are further demonstrated by the small interfering RNA (siRNA)-TREM2 transfected BV2 cells, which are obviously not influenced by CSPQ nanoparticles. The cell membrane coated CSPQ (referred as CSPQ@CM) nanoparticles can significantly reduce sevoflurane-induced learning and memory deficits, improve lipid metabolism dysfunction, and promote the remyelination in the hippocampus of mice. The study shows great potential of targeting microglial lipid metabolism in promoting remyelination of neurons for treatment of neurotoxicity and neurodegenerative diseases.
Collapse
Affiliation(s)
- Wenting Li
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical CollegeSoochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Xiaowen Meng
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Ke Peng
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Yaobao Han
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical CollegeSoochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123China
| | - Hanghang Liu
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical CollegeSoochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123China
| | - Weiming Zhao
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Gang Wang
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical CollegeSoochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Li Deng
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| | - Hong Liu
- Department of Anaesthesiology and Pain MedicineUniversity of California Davis HealthSacramentoCA 95817USA
| | - Zhen Li
- Center for Molecular Imaging and Nuclear MedicineState Key Laboratory of Radiation Medicine and ProtectionSchool for Radiological and Interdisciplinary Sciences (RAD‐X)Suzhou Medical CollegeSoochow UniversityCollaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education InstitutionsSuzhou215123China
| | - Fuhai Ji
- Department of Anesthesiologythe First Affiliated Hospital of Soochow UniversitySuzhouJiangsu215006China
- Institute of AnesthesiologySoochow UniversitySuzhouJiangsu215006China
| |
Collapse
|
48
|
Franklin RJM, Bodini B, Goldman SA. Remyelination in the Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041371. [PMID: 38316552 PMCID: PMC10910446 DOI: 10.1101/cshperspect.a041371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
49
|
Lattau SSJ, Borsch LM, Auf dem Brinke K, Klose C, Vinhoven L, Nietert M, Fitzner D. Plasma Lipidomic Profiling Using Mass Spectrometry for Multiple Sclerosis Diagnosis and Disease Activity Stratification (LipidMS). Int J Mol Sci 2024; 25:2483. [PMID: 38473733 DOI: 10.3390/ijms25052483] [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: 01/16/2024] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
This investigation explores the potential of plasma lipidomic signatures for aiding in the diagnosis of Multiple Sclerosis (MS) and evaluating the clinical course and disease activity of diseased patients. Plasma samples from 60 patients with MS (PwMS) were clinically stratified to either a relapsing-remitting (RRMS) or a chronic progressive MS course and 60 age-matched controls were analyzed using state-of-the-art direct infusion quantitative shotgun lipidomics. To account for potential confounders, data were filtered for age and BMI correlations. The statistical analysis employed supervised and unsupervised multivariate data analysis techniques, including a principal component analysis (PCA), a partial least squares discriminant analysis (oPLS-DA) and a random forest (RF). To determine whether the significant absolute differences in the lipid subspecies have a relevant effect on the overall composition of the respective lipid classes, we introduce a class composition visualization (CCV). We identified 670 lipids across 16 classes. PwMS showed a significant increase in diacylglycerols (DAG), with DAG 16:0;0_18:1;0 being proven to be the lipid with the highest predictive ability for MS as determined by RF. The alterations in the phosphatidylethanolamines (PE) were mainly linked to RRMS while the alterations in the ether-bound PEs (PE O-) were found in chronic progressive MS. The amount of CE species was reduced in the CPMS cohort whereas TAG species were reduced in the RRMS patients, both lipid classes being relevant in lipid storage. Combining the above mentioned data analyses, distinct lipidomic signatures were isolated and shown to be correlated with clinical phenotypes. Our study suggests that specific plasma lipid profiles are not merely associated with the diagnosis of MS but instead point toward distinct clinical features in the individual patient paving the way for personalized therapy and an enhanced understanding of MS pathology.
Collapse
Affiliation(s)
| | - Lisa-Marie Borsch
- Department of Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | | | | | - Liza Vinhoven
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Manuel Nietert
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Dirk Fitzner
- Department of Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
50
|
Sun R, Lei C, Xu Z, Gu X, Huang L, Chen L, Tan Y, Peng M, Yaddanapudi K, Siskind L, Kong M, Mitchell R, Yan J, Deng Z. Neutral ceramidase regulates breast cancer progression by metabolic programming of TREM2-associated macrophages. Nat Commun 2024; 15:966. [PMID: 38302493 PMCID: PMC10834982 DOI: 10.1038/s41467-024-45084-7] [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: 02/12/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
The tumor microenvironment is reprogrammed by cancer cells and participates in all stages of tumor progression. Neutral ceramidase is a key regulator of ceramide, the central intermediate in sphingolipid metabolism. The contribution of neutral ceramidase to the reprogramming of the tumor microenvironment is not well understood. Here, we find that deletion of neutral ceramidase in multiple breast cancer models in female mice accelerates tumor growth. Our result show that Ly6C+CD39+ tumor-infiltrating CD8 T cells are enriched in the tumor microenvironment and display an exhausted phenotype. Deletion of myeloid neutral ceramidase in vivo and in vitro induces exhaustion in tumor-infiltrating Ly6C+CD39+CD8+ T cells. Mechanistically, myeloid neutral ceramidase is required for the generation of lipid droplets and for the induction of lipolysis, which generate fatty acids for fatty-acid oxidation and orchestrate macrophage metabolism. Metabolite ceramide leads to reprogramming of macrophages toward immune suppressive TREM2+ tumor associated macrophages, which promote CD8 T cells exhaustion.
Collapse
Affiliation(s)
- Rui Sun
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Chao Lei
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Zhishan Xu
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Xuemei Gu
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Liu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, P.R. China
| | - Liang Chen
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Yi Tan
- Department of Pediatrics and Pediatric Research Institute, University of Louisville, Louisville, KY, USA
| | - Min Peng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Kavitha Yaddanapudi
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Leah Siskind
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Maiying Kong
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Robert Mitchell
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Jun Yan
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA
| | - Zhongbin Deng
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA.
- Brown Cancer Center, University of Louisville, Louisville, KY, KY40202, USA.
| |
Collapse
|