1
|
Chen Y, Wei Y, Liu J, Zhu T, Zhou C, Zhang D. Spatial transcriptomics combined with single-nucleus RNA sequencing reveals glial cell heterogeneity in the human spinal cord. Neural Regen Res 2025; 20:3302-3316. [PMID: 38934400 PMCID: PMC11881709 DOI: 10.4103/nrr.nrr-d-23-01876] [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: 11/14/2023] [Revised: 03/04/2024] [Accepted: 04/30/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00032/figure1/v/2024-12-20T164640Z/r/image-tiff Glial cells play crucial roles in regulating physiological and pathological functions, including sensation, the response to infection and acute injury, and chronic neurodegenerative disorders. Glial cells include astrocytes, microglia, and oligodendrocytes in the central nervous system, and satellite glial cells and Schwann cells in the peripheral nervous system. Despite the greater understanding of glial cell types and functional heterogeneity achieved through single-cell and single-nucleus RNA sequencing in animal models, few studies have investigated the transcriptomic profiles of glial cells in the human spinal cord. Here, we used high-throughput single-nucleus RNA sequencing and spatial transcriptomics to map the cellular and molecular heterogeneity of astrocytes, microglia, and oligodendrocytes in the human spinal cord. To explore the conservation and divergence across species, we compared these findings with those from mice. In the human spinal cord, astrocytes, microglia, and oligodendrocytes were each divided into six distinct transcriptomic subclusters. In the mouse spinal cord, astrocytes, microglia, and oligodendrocytes were divided into five, four, and five distinct transcriptomic subclusters, respectively. The comparative results revealed substantial heterogeneity in all glial cell types between humans and mice. Additionally, we detected sex differences in gene expression in human spinal cord glial cells. Specifically, in all astrocyte subtypes, the levels of NEAT1 and CHI3L1 were higher in males than in females, whereas the levels of CST3 were lower in males than in females. In all microglial subtypes, all differentially expressed genes were located on the sex chromosomes. In addition to sex-specific gene differences, the levels of MT-ND4 , MT2A , MT-ATP6 , MT-CO3 , MT-ND2 , MT-ND3 , and MT-CO2 in all spinal cord oligodendrocyte subtypes were higher in females than in males. Collectively, the present dataset extensively characterizes glial cell heterogeneity and offers a valuable resource for exploring the cellular basis of spinal cord-related illnesses, including chronic pain, amyotrophic lateral sclerosis, and multiple sclerosis.
Collapse
Affiliation(s)
- Yali Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Ning W, Lv S, Wang Q, Xu Y. The pivotal role of microglia in injury and the prognosis of subarachnoid hemorrhage. Neural Regen Res 2025; 20:1829-1848. [PMID: 38993136 PMCID: PMC11691474 DOI: 10.4103/nrr.nrr-d-24-00241] [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: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/25/2024] [Indexed: 07/13/2024] Open
Abstract
Subarachnoid hemorrhage leads to a series of pathological changes, including vascular spasm, cellular apoptosis, blood-brain barrier damage, cerebral edema, and white matter injury. Microglia, which are the key immune cells in the central nervous system, maintain homeostasis in the neural environment, support neurons, mediate apoptosis, participate in immune regulation, and have neuroprotective effects. Increasing evidence has shown that microglia play a pivotal role in the pathogenesis of subarachnoid hemorrhage and affect the process of injury and the prognosis of subarachnoid hemorrhage. Moreover, microglia play certain neuroprotective roles in the recovery phase of subarachnoid hemorrhage. Several approaches aimed at modulating microglia function are believed to attenuate subarachnoid hemorrhage injury. This provides new targets and ideas for the treatment of subarachnoid hemorrhage. However, an in-depth and comprehensive summary of the role of microglia after subarachnoid hemorrhage is still lacking. This review describes the activation of microglia after subarachnoid hemorrhage and their roles in the pathological processes of vasospasm, neuroinflammation, neuronal apoptosis, blood-brain barrier disruption, cerebral edema, and cerebral white matter lesions. It also discusses the neuroprotective roles of microglia during recovery from subarachnoid hemorrhage and therapeutic advances aimed at modulating microglial function after subarachnoid hemorrhage. Currently, microglia in subarachnoid hemorrhage are targeted with TLR inhibitors, nuclear factor-κB and STAT3 pathway inhibitors, glycine/tyrosine kinases, NLRP3 signaling pathway inhibitors, Gasdermin D inhibitors, vincristine receptor α receptor agonists, ferroptosis inhibitors, genetic modification techniques, stem cell therapies, and traditional Chinese medicine. However, most of these are still being evaluated at the laboratory stage. More clinical studies and data on subarachnoid hemorrhage are required to improve the treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Wenjing Ning
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Shi Lv
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| |
Collapse
|
3
|
Panda P, Mohanty S, Gouda SR, Mohapatra R. Advances in nanomedicine for retinal drug delivery: overcoming barriers and enhancing therapeutic outcomes. J Drug Target 2025; 33:587-611. [PMID: 39694681 DOI: 10.1080/1061186x.2024.2443144] [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: 10/22/2024] [Revised: 11/16/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Nanomedicine offers a promising avenue for improving retinal drug delivery, effectively addressing challenges associated with ocular diseases like age-related macular degeneration and diabetic retinopathy. Nanoparticles, with their submicron size and customisable surface properties, enable enhanced permeability and retention within retinal tissues, supporting sustained drug release and minimising systemic side effects. Nanostructured scaffolds further provide a supportive environment for retinal cell growth and tissue regeneration, crucial for treating degenerative conditions. Additionally, advanced nanodevices facilitate real-time monitoring and controlled drug release, marking significant progress in retinal therapy. This study reviews recent advancements in nanomedicine for retinal drug delivery, critically analysing design innovations, therapeutic benefits, and limitations of these systems. By advancing nanotechnology integration in ocular therapies, this field holds strong potential for overcoming current barriers, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Pratikeswar Panda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Shreyashree Mohanty
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Sangita Ranee Gouda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Rajaram Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| |
Collapse
|
4
|
Kazemi M, Sanati M, Shekari Khaniani M, Ghafouri-Fard S. A review on the lncRNA-miRNA-mRNA regulatory networks involved in inflammatory processes in Alzheimer's disease. Brain Res 2025; 1856:149595. [PMID: 40132722 DOI: 10.1016/j.brainres.2025.149595] [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: 01/22/2025] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Alzheimer's disease is a progressive neurodegenerative condition that is the most frequent reason for dementia. Due to the increasing trend of aging in societies, it will place a large social and financial burden on society. Although beta amyloid plaques and the formation of neurofibrillary tangles are mentioned as the main events in this disorder, the exact molecular pathology and inflammatory regulatory networks involved in neuroinflammatory events, as a fundamental pathogenic mechanism remain unknown. Understanding these molecular network pathways in addition to helping to understand the pathogenesis of Alzheimer's disease, can also help in the early diagnosis as well as the control of inflammatory processes that are involved in its progression. So, in this study, we intend to have an overview on the regulatory lncRNAs of Alzheimer's disease and their related miRNA and mRNAs, as well as the relationship of these regulatory pathways with inflammatory processes, so that we can provide a perspective for future studies in the field of diagnosis and possibly treatment of this disorder.
Collapse
Affiliation(s)
- Masoumeh Kazemi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahla Sanati
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Shekari Khaniani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Zhu Y, Li X, Lei X, Tang L, Wen D, Zeng B, Zhang X, Huang Z, Guo Z. The potential mechanism and clinical application value of remote ischemic conditioning in stroke. Neural Regen Res 2025; 20:1613-1627. [PMID: 38845225 PMCID: PMC11688546 DOI: 10.4103/nrr.nrr-d-23-01800] [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: 11/02/2023] [Revised: 01/30/2024] [Accepted: 03/19/2024] [Indexed: 08/07/2024] Open
Abstract
Some studies have confirmed the neuroprotective effect of remote ischemic conditioning against stroke. Although numerous animal researches have shown that the neuroprotective effect of remote ischemic conditioning may be related to neuroinflammation, cellular immunity, apoptosis, and autophagy, the exact underlying molecular mechanisms are unclear. This review summarizes the current status of different types of remote ischemic conditioning methods in animal and clinical studies and analyzes their commonalities and differences in neuroprotective mechanisms and signaling pathways. Remote ischemic conditioning has emerged as a potential therapeutic approach for improving stroke-induced brain injury owing to its simplicity, non-invasiveness, safety, and patient tolerability. Different forms of remote ischemic conditioning exhibit distinct intervention patterns, timing, and application range. Mechanistically, remote ischemic conditioning can exert neuroprotective effects by activating the Notch1/phosphatidylinositol 3-kinase/Akt signaling pathway, improving cerebral perfusion, suppressing neuroinflammation, inhibiting cell apoptosis, activating autophagy, and promoting neural regeneration. While remote ischemic conditioning has shown potential in improving stroke outcomes, its full clinical translation has not yet been achieved.
Collapse
Affiliation(s)
- Yajun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoguo Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingwei Lei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuyang Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daochen Wen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Zeng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofeng Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zichao Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zongduo Guo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Wan F, Zhuang K, Li Z, Wang X, Li W, Hou Y, You W, Jiang Y, Wang M, Zhu P. Electroacupuncture mitigates cognitive impairments in chronic hypoxia-induced mice by modulating neuroinflammation. IBRO Neurosci Rep 2025; 18:432-442. [PMID: 40144798 PMCID: PMC11937942 DOI: 10.1016/j.ibneur.2025.03.001] [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: 11/11/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigates the therapeutic effects and mechanisms of electroacupuncture (EA) on cognitive impairment induced by chronic hypoxia (CH) in mice. Chronic hypoxia was simulated by exposing mice to a 10 % oxygen environment for 8 hours daily over 3 months. The cognitive functions of the mice were assessed through behavioral tests, including the open field test (OFT), Y-maze, and Morris water maze (MWM). Results showed that CH induced significant anxiety-like behaviors and memory impairments in mice. EA treatment, targeting the Baihui (GV20), Shenting (GV24), and Zusanli (ST36) acupoints, significantly ameliorated these behavioral deficits. Histological analyses using HE staining, Nissl staining, and TUNEL assays demonstrated that EA reduced neuronal damage, apoptosis, and myelin loss in the cerebral cortex and hippocampus. Additionally, EA treatment significantly lowered the expression of the pro-inflammatory cytokine TNF-α in brain tissues, suggesting its anti-inflammatory effects. Immunofluorescence and Western blot analyses revealed that EA inhibited the overactivation of microglia and astrocytes in the CH model. Specifically, EA suppressed the expression of Iba1 and GFAP, markers of microglial and astrocytic activation, respectively. Furthermore, EA promoted the polarization of microglia towards the M2 anti-inflammatory phenotype by downregulating iNOS and upregulating Arg1. Similarly, EA reduced the expression of C3, a marker of A1 astrocytes, thereby preventing astrocytic polarization towards the pro-inflammatory state. Organotypic brain slice cultures subjected to oxygen-glucose deprivation (OGD) confirmed that electrical stimulation, akin to EA, inhibited the activation of microglia and astrocytes under hypoxic conditions. In conclusion, EA improves cognitive function in CH-induced mice by reducing neuroinflammation, inhibiting glial cell overactivation, and promoting anti-inflammatory phenotypes. These findings highlight EA's potential as a therapeutic intervention for cognitive impairments related to chronic hypoxia.
Collapse
Affiliation(s)
- Fang Wan
- International Acupuncture and Moxibustion Innovation Institute, School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Kun Zhuang
- Dongguan Integrated Traditional Chinese and Western Medicine Hospital, Dongguan 523819, China
| | - Ziyu Li
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xiaoqing Wang
- Chinese Medicine Research Institute, Chengde Medical University, Chengde 067000, China
| | - Wenyan Li
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yunlong Hou
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang Yiling Pharmaceutical Co. Ltd., Shijiazhuang 050000, China
- Key Laboratory of State Administration of TCM (Cardio-Cerebral VesselCollateral Disease), Shijiazhuang 050000, China
| | - Wanhui You
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Yibing Jiang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mingye Wang
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Pengyu Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Sharma P, Daksh R, Khanna S, Mudgal J, Lewis SA, Arora D, Nampoothiri M. Microglial cannabinoid receptor 2 and epigenetic regulation: Implications for the treatment of depression. Eur J Pharmacol 2025; 995:177422. [PMID: 39988094 DOI: 10.1016/j.ejphar.2025.177422] [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: 10/07/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Depression, often stress-induced, is closely related to neuroinflammation, in which microglia, the brain's immune cells, are the leading players. Microglia shift between a quiescent and an active state, promoting both pro- and anti-inflammatory responses. Cannabinoid type 2 (CB2) receptor encoded by the CNR2 gene is a key player to modulate inflammatory activity. CB2 receptor is highly controlled at the epigenetic level, especially in response to stressful stimuli, positioning it between stress, neuroinflammation, and depression. The following review addresses how epigenetic regulation of CNR2 expression affects depression and the dissection, further, of molecular pathways driving neuroinflammation-related depressive states. The present study emphasizes the therapeutic potential of CB2 receptor agonists that selectively interact with activated microglia and opens a new avenue for the treatment of depression associated with neuroinflammation. The review, therefore, provides a framework of underlying mechanisms for developing novel therapeutic strategies that focus on relieving symptoms by modulating the neuroinflammatory response. Finally, this review underlines the possibilities of therapeutic interventions taking into account CB2 receptors in combating depression.
Collapse
Affiliation(s)
- Pratyasha Sharma
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Rajni Daksh
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Saumya Khanna
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Devinder Arora
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, 4222, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
8
|
Kumar S, Kahle AD, Keeler AB, Zunder ER, Deppmann CD. Characterizing Microglial Signaling Dynamics During Inflammation Using Single-Cell Mass Cytometry. Glia 2025; 73:1022-1035. [PMID: 39780484 PMCID: PMC11920681 DOI: 10.1002/glia.24670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Microglia play a critical role in maintaining central nervous system (CNS) homeostasis and display remarkable plasticity in their response to inflammatory stimuli. However, the specific signaling profiles that microglia adopt during such challenges remain incompletely understood. Traditional transcriptomic approaches provide valuable insights, but fail to capture dynamic post-translational changes. In this study, we utilized time-resolved single-cell mass cytometry (CyTOF) to measure distinct signaling pathways activated in microglia upon exposure to bacterial and viral mimetics-lipopolysaccharide (LPS) and polyinosinic-polycytidylic acid (Poly(I:C)), respectively. Furthermore, we evaluated the immunomodulatory role of astrocytes on microglial signaling in mixed cultures. Microglia or mixed cultures derived from neonatal mice were treated with LPS or Poly(I:C) for 48 h. Cultures were stained with a panel of 33 metal-conjugated antibodies targeting signaling and identity markers. High-dimensional clustering analysis was used to identify emergent signaling modules. We found that LPS treatment led to more robust early activation of pp38, pERK, pRSK, and pCREB compared to Poly(I:C). Despite these differences, both LPS and Poly(I:C) upregulated the classical reactivity markers CD40 and CD86 at later time points. Strikingly, the presence of astrocytes significantly blunted microglial responses to both stimuli, particularly dampening CD40 upregulation. Our studies demonstrate that single-cell mass cytometry effectively captures the dynamic signaling landscape of microglia under pro-inflammatory conditions. This approach may pave the way for targeted therapeutic investigations of various neuroinflammatory disorders. Moreover, our findings underscore the necessity of considering cellular context, such as astrocyte presence, in interpreting microglial behavior during inflammation.
Collapse
Affiliation(s)
- Sushanth Kumar
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - August D Kahle
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Austin B Keeler
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Eli R Zunder
- Department of Biomedical Engineering, School of Engineering, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Christopher D Deppmann
- Department of Biology, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, School of Engineering, University of Virginia, Charlottesville, Virginia, USA
- Program in Fundamental Neuroscience, College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
9
|
Tang X, Huang L, Ma W, Huang M, Zeng Z, Yu Y, Qin N, Zhou F, Li F, Gong S, Yang H. Intestinal 8 gingerol attenuates TBI-induced neuroinflammation by inhibiting microglia NLRP3 inflammasome activation in a PINK1/Parkin-dependent manner. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156580. [PMID: 40058316 DOI: 10.1016/j.phymed.2025.156580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/23/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND traumatic brain injury (TBI) is irreversible brain damage, leading to inflammation and cognitive dysfunction. Microglia involved in the inflammatory response after TBI. The gut microbiota, known as the body's "second brain," regulates neurogenesis and immune responses, but its precise role in regulating TBI remains unclear. PURPOSE to investigate the effect of gut microbiota and metabolites disorder on TBI injury. STUDY DESIGN 16SrRNA and metabolomics compared gut microbiota and metabolites in sham group and TBI group, then proved that the differential metabolite 8-gingerol (8G) alleviated the microglia neuroinflammatory response after TBI. METHODS fecal microbiota transplantation explored the role of dysbiosis in TBI. LC/MS detected the content of 8-gingerol in cecum, blood, and brain. HE, Nissl, Tunel staining and mNSS score evaluated brain injury. Western blot and immunofluorescence detected the expression of inflammasome-related proteins and mitophagy-related proteins in brain tissue and BV2 cells. RNA sequencing analyzed the molecular mechanism of 8-gingerol. RESULT rats transplanted with TBI feces had worse brain injury and neurological deficits than those with normal feces. 16SrRNA and metabolomics found that TBI caused dysbiosis and decreased 8-gingerol level, leading to severe neuroinflammation. Mechanistically, 8-gingerol inhibited NLRP3 inflammasome by promoting PINK1-Parkin mediated mitophagy in microglia. Inhibition of Parkin, through either small interfering RNA or the inhibitor 3MA reversed the inhibitory effect of 8-gingerol on NLRP3 by blocking mitophagy. BV2 cells transcriptome showed that 8-gingerol significantly increased the expression of autophagy factor Wipi1, and small interfering RNA of Wipi1 abolished the effect of 8-gingerol on promoting mitophagy and the inhibitory effect on NLRP3. CONCLUSION our findings shed light on the pivotal role of gut microbes in TBI, and identify 8 gingerol as an important anti-inflammatory compound during TBI.
Collapse
Affiliation(s)
- Xuheng Tang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Lin Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Weiquan Ma
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Mingxin Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yiqin Yu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Na Qin
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China
| | - Fei Zhou
- Central Hospital of Guangdong Prison, Guangzhou 510430, China
| | - Fen Li
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China.
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, 510515, China.
| | - Hong Yang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, 510665, China; The Third Clinical College of Southern Medical University, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China.
| |
Collapse
|
10
|
Xue F, Zhang M, Zhao RY, Wang XW, Gu Y, Yang Y, Chen WF. Dectin-1 participates in neuroinflammation and dopaminergic neurodegeneration through synergistic signaling crosstalk with TLR4. Brain Behav Immun 2025; 126:260-273. [PMID: 39978698 DOI: 10.1016/j.bbi.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Neuroinflammation mediated by microglial activation plays a prominent role in the pathogenesis of Parkinson's disease (PD). Dendritic cell-associated C-type lectin-1 (Dectin-1) is a pattern recognition receptor that is involved in innate immunity. However, the role of Dectin-1 on dopaminergic neuronal damage remains unclear. Our results demonstrated that the expression of Dectin-1 was significantly increased in the microglia of the LPS-induced PD mouse model. Inhibition of Dectin-1 by laminarin (LAM) attenuated LPS-induced dopaminergic neuronal damage in substantia nigra (SN) and behavioral deficits and promoted the phenotypic transformation of microglia from M1 to M2. Moreover, inhibition or knockdown of Dectin-1 significantly decreased LPS-induced phosphorylation of Syk and P65 as well as the production of COX-2 and iNOS in BV2 cells. Knockdown of Syk also significantly decreased LPS-induced protein expressions of COX-2 and iNOS. Mechanistically, both TLR4 inhibitor and NF-κB inhibitor could antagonize LPS-induced Dectin-1 expression. Chromatin immunoprecipitation (ChIP) assays showed a physical binding of NF-κB/P65 to Dectin-1 promoter, which further indicated the regulatory effect of toll-like receptor 4 (TLR4)/NF-κB signaling pathway on Dectin-1 expression. Furthermore, the present study provided the first evidence that Dectin-1 activation by hot-alkali treated depleted zymosan (d-Zymosan) could induce dopaminergic neurotoxicity and motor dysfunction, and promote up-regulation of TLR4, iNOS and Iba-1 in C57BL/6J mice. In conclusion, Dectin-1-Syk synergistic signaling crosstalk with TLR4/NF-κB promotes and maintains inflammatory phenotypes of M1 microglia which induces dopaminergic neuronal damage in SN. These findings provide novel insights into the pivotal role of Dectin-1 in neuroinflammation, suggesting its potential as a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Feng Xue
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mei Zhang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Rui-Yue Zhao
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Xiao-Wen Wang
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yu Gu
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Ye Yang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, China
| | - Wen-Fang Chen
- Department of Physiology, Shandong Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
11
|
Liu L, Xiao F, Yang J, Yao H, Hua K. Microglial pyroptosis induced by SENP7 via the cGAS/STING/IRF3 pathway contributes to neuronal apoptosis. Cytokine 2025; 189:156893. [PMID: 39999677 DOI: 10.1016/j.cyto.2025.156893] [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: 12/10/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Maternal anesthetic exposure may exacerbate significant neurocognitive risks in the immature brains of fetuses. However, the mechanisms through which sevoflurane exposure during pregnancy results in cognitive impairments in offspring remain unclear. METHODS Pregnant C57BL/6 mice (gestational day 14) were intervented with 2.5 % sevoflurane for 6 h. Morris water maze test and context fear conditioning test were utilized to evaluate the cognitive function of the offspring. BV2 cells were stimulated with LPS-ATP to evaluate the impacts of SENP7 on microglial pyroptosis. A co-culture experiment was conducted to investigate the apoptosis of mouse hippocampal neuronal cells induced by BV2 cells. The regulatory roles of SENP7 in the cGAS/STING/IRF3 pathway were assessed using an immunoprecipitation SUMOylation assay, along with Western blot analysis. RESULTS Sevoflurane exposure during pregnancy resulted in cognitive impairments in offspring mice, which were associated with the upregulation of SENP7, Iba1, Caspase1, and GSDMD-N proteins, as well as the downregulation of NeuN and TH proteins in the brains of the offspring. The knockdown of SENP7 inhibited the elevation of GSDMD-N, Caspase1, and NLRP3 protein levels, subsequently reducing the concentrations of IL-1β and IL-18 in BV2 cells induced by LPS-ATP. Furthermore, SENP7 facilitated the activation of the cGAS/STING/IRF3 axis by regulating the deSUMOylation of cGAS, which triggered microglial pyroptosis and subsequently led to neuronal apoptosis. CONCLUSION Maternal exposure to sevoflurane increased the expression of SENP7 in the brains of offspring and resulted in detrimental effects on cognitive function. This phenomenon was associated with neuronal apoptosis triggered by microglial pyroptosis, which was regulated by SENP7 through the cGAS/STING/IRF3 pathway.
Collapse
Affiliation(s)
- Lin Liu
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing Women and Children's Hospital, Wenzhou Medical University, Jiaxing 314050, Zhejiang, PR China
| | - Fei Xiao
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing Women and Children's Hospital, Wenzhou Medical University, Jiaxing 314050, Zhejiang, PR China
| | - Jinyue Yang
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing Women and Children's Hospital, Wenzhou Medical University, Jiaxing 314050, Zhejiang, PR China.
| | - Hanqing Yao
- Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Jiaxing Women and Children's Hospital, Wenzhou Medical University, Jiaxing 314050, Zhejiang, PR China
| | - Ke Hua
- Department of Reproductive Medicine Center, Jiaxing Maternity and Child Health Care Hospital, Jiaxing Women and Children's Hospital, Wenzhou Medical University, Jiaxing 314050, Zhejiang, PR China
| |
Collapse
|
12
|
Cao H, Liu T, Xu M. Senkyunolide I Improves Septicemia-Induced Brain Dysfunction via Regulating Nrf2 and Astrocyte Activity. Biotechnol Appl Biochem 2025:e2748. [PMID: 40229948 DOI: 10.1002/bab.2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/23/2024] [Indexed: 04/16/2025]
Abstract
Senkyunolide I (Sen I) has a protective effect on the blood-brain barrier (BBB) in rats with sepsis-associated encephalopathy (SAE). This study investigated whether Sen I regulates Nrf2 to ameliorate sepsis-induced brain dysfunction (SIBD). Sixty rats were randomly assigned into Sham group, SAE group (Model group), SAE + Sen I group (72 mg/kg, Sen I group), and SAE+ positive control group (RTA 402, Nrf2 receptor agonist, RTA 402 group), with 15 rats in each group. The cecal ligation and puncture (CLP) method was applied to induce sepsis in rats. SAE modeling was verified 6 h after operation. The drug was administered 24 h after surgery. Six rats in each group were sacrificed 24 h after administration, with brains extracted. The remaining rats would continue to be observed for their survival status until 72 h post-surgery. Brain cell apoptosis was measured using TUNEL. We detected the expression of glial fibrillary acidic protein (GFAP) by immunofluorescence, Nrf2 gene expression by RT-qPCR, and the protein expression of Nrf2, MMP-9, AQP-4, and occludin by Western blot. TNF-α and IL-1β levels were tested by ELISA, and malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) by biochemical tests. Survival rate at 72 h post-surgery, Sham group was 100%. The survival rate of the Sen I group (44.4%) and the RTA 402 group (55.6%) is significantly higher than that of the Model group (11.1%). Both Sen I and RTA 402 can improve the brain tissue damage in rats caused by sepsis, specifically by reducing apoptosis and GFAP expression, reducing TNF-α, IL-1β, and MDA levels, increasing the activity of GSH-Px, downregulating the protein expression of MMP-9 and AQP-4, and upregulating the protein expression of Nrf2 and occludin. Moreover, Sen I significantly increased the expression of Nrf2 in rat brain tissues. Sen I ameliorates SIBD in rats by regulating the expression of Nrf2 and astrocyte activation.
Collapse
Affiliation(s)
- Haohao Cao
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei, China
| | - Tao Liu
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei, China
| | - Meixia Xu
- Department of Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, Hubei, China
| |
Collapse
|
13
|
Garton T, Smith MD, Kesharwani A, Gharagozloo M, Oh S, Na CH, Absinta M, Reich DS, Zack DJ, Calabresi PA. Myeloid lineage C3 induces reactive gliosis and neuronal stress during CNS inflammation. Nat Commun 2025; 16:3481. [PMID: 40216817 PMCID: PMC11992029 DOI: 10.1038/s41467-025-58708-3] [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: 08/04/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Complement component C3 mediates pathology in CNS neurodegenerative diseases. Here we use scRNAseq of sorted C3-reporter positive cells from mouse brain and optic nerve to characterize C3 producing glia in experimental autoimmune encephalomyelitis (EAE), a model in which peripheral immune cells infiltrate the CNS, causing reactive gliosis and neuro-axonal pathology. We find that C3 expression in the early inflammatory stage of EAE defines disease-associated glial subtypes characterized by increased expression of genes associated with mTOR activation and cell metabolism. This pro-inflammatory subtype is abrogated with genetic C3 depletion, a finding confirmed with proteomic analyses. In addition, early optic nerve axonal injury and retinal ganglion cell oxidative stress, but not loss of post-synaptic density protein 95, are ameliorated by selective deletion of C3 in myeloid cells. These data suggest that in addition to C3b opsonization of post synaptic proteins leading to neuronal demise, C3 activation is a contributor to reactive glia in the optic nerve.
Collapse
Affiliation(s)
- Thomas Garton
- Department of Neurology, 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
| | - Ajay Kesharwani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sungtaek Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Chan-Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Reserach Hospital, Milan, Italy
| | - Daniel S Reich
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
14
|
Dali R, Langlet F. Tanycytes in the Nexus of Hypothalamic Inflammation, Appetite Control, and Obesity. Physiol Behav 2025:114917. [PMID: 40222438 DOI: 10.1016/j.physbeh.2025.114917] [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: 12/30/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Hypothalamic inflammation has been identified as a critical factor driving the development of obesity and associated metabolic disorders. This inflammation-related disruption of energy balance relies on alterations in metabolic cues sensing and hypothalamic cellular functions, together leading to overeating and weight gain. Within the hypothalamic cellular networks controlling energy balance, recent studies have highlighted the significance of glial dysfunction in these processes, suggesting that these cells could provide new avenues for weight loss therapies. Glia rapidly activates following the consumption of a high-fat diet, even after a very short exposure, and contributes to the disruption of the entire system through inflammatory crosstalk. This review explores recent progress in understanding the molecular interactions between glial cells and neurons in hypothalamic inflammation related to obesity, diabetes, and associated complications. Notably, it highlights specialized ependymal cells called tanycytes, whose role is still underestimated in hypothalamic inflammation, and examines the potential for targeting this cell type as a treatment strategy for metabolic disorders.
Collapse
Affiliation(s)
- Rafik Dali
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
15
|
Kimura K, Subramanian A, Yin Z, Khalilnezhad A, Wu Y, He D, Dixon KO, Chitta UK, Ding X, Adhikari N, Guzchenko I, Zhang X, Tang R, Pertel T, Myers SA, Aastha A, Nomura M, Eskandari-Sedighi G, Singh V, Liu L, Lambden C, Kleemann KL, Gupta N, Barry JL, Durao A, Cheng Y, Silveira S, Zhang H, Suhail A, Delorey T, Rozenblatt-Rosen O, Freeman GJ, Selkoe DJ, Weiner HL, Blurton-Jones M, Cruchaga C, Regev A, Suvà ML, Butovsky O, Kuchroo VK. Immune checkpoint TIM-3 regulates microglia and Alzheimer's disease. Nature 2025:10.1038/s41586-025-08852-z. [PMID: 40205047 DOI: 10.1038/s41586-025-08852-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 02/28/2025] [Indexed: 04/11/2025]
Abstract
Microglia are the resident immune cells in the brain and have pivotal roles in neurodevelopment and neuroinflammation1,2. This study investigates the function of the immune-checkpoint molecule TIM-3 (encoded by HAVCR2) in microglia. TIM-3 was recently identified as a genetic risk factor for late-onset Alzheimer's disease3, and it can induce T cell exhaustion4. However, its specific function in brain microglia remains unclear. We demonstrate in mouse models that TGFβ signalling induces TIM-3 expression in microglia. In turn, TIM-3 interacts with SMAD2 and TGFBR2 through its carboxy-terminal tail, which enhances TGFβ signalling by promoting TGFBR-mediated SMAD2 phosphorylation, and this process maintains microglial homeostasis. Genetic deletion of Havcr2 in microglia leads to increased phagocytic activity and a gene-expression profile consistent with the neurodegenerative microglial phenotype (MGnD), also referred to as disease-associated microglia (DAM). Furthermore, microglia-targeted deletion of Havcr2 ameliorates cognitive impairment and reduces amyloid-β pathology in 5×FAD mice (a transgenic model of Alzheimer's disease). Single-nucleus RNA sequencing revealed a subpopulation of MGnD microglia in Havcr2-deficient 5×FAD mice characterized by increased pro-phagocytic and anti-inflammatory gene expression alongside reduced pro-inflammatory gene expression. These transcriptomic changes were corroborated by single-cell RNA sequencing data across most microglial clusters in Havcr2-deficient 5×FAD mice. Our findings reveal that TIM-3 mediates microglia homeostasis through TGFβ signalling and highlight the therapeutic potential of targeting microglial TIM-3 in Alzheimer's disease.
Collapse
Affiliation(s)
- Kimitoshi Kimura
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ayshwarya Subramanian
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhuoran Yin
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ahad Khalilnezhad
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yufan Wu
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Danyang He
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Karen O Dixon
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Udbhav Kasyap Chitta
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiaokai Ding
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Niraj Adhikari
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Isabell Guzchenko
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xiaoming Zhang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ruihan Tang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas Pertel
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Samuel A Myers
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Aastha Aastha
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Masashi Nomura
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ghazaleh Eskandari-Sedighi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | | | - Lei Liu
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Conner Lambden
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kilian L Kleemann
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Neha Gupta
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jen-Li Barry
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana Durao
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yiran Cheng
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sebastian Silveira
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Huiyuan Zhang
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aamir Suhail
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni Delorey
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St Louis, St Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St Louis, MO, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Mario L Suvà
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Vijay K Kuchroo
- The Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
Cai Y, Lin Z, Shen X, Li M, Xing L, Yang T, Chen G. Effect of microglial Pd1 on glial scar formation after spinal cord injury in mice. J Biol Chem 2025:108489. [PMID: 40209954 DOI: 10.1016/j.jbc.2025.108489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The crosstalk between microglia and astrocytes following spinal cord injury (SCI) greatly decides the prognosis. However, a comprehensive understanding of the molecular mechanisms by which microglia regulate astrocytic activity post-SCI is lacking. Programmed cell death protein 1 (Pdcd1, Pd1) plays a crucial role in modulating immune responses by exerting suppressive effects on microglia and peripheral immune cells within the central nervous system (CNS). Previous studies have shown the involvement of Pd1 in the pathogenesis of SCI; however, the role of microglial Pd1 in astrocytic activation and the following glial scar formation remains elusive. Here, we demonstrated that the pharmacological depletion of microglia using minocycline decreased the expression of TNF-α and IL-6 while concurrently increasing the expression of IL-10 following SCI, thereby facilitating motor function recovery in mice. We observed an increase in Pd1 expression in the injured spinal cord after SCI, with precise localization of Pd1 within microglia. Based on Pd1 knockout (KO) mice, we further revealed that Pd1 deficiency disrupted glial scar formation, leading to increased inflammation, impeded nerve regeneration, enlarged tissue damage, and compromised functional recovery following SCI. In vitro study showed that siRNA-mediated inhibition of Pd1 in microglia followed by lipopolysaccharide (LPS) treatment significantly inhibited astrocyte migration and upregulated the secretion of TNF-α and CXCL9 from microglia, indicating that microglial Pd1 regulates glial scar formation through modulating the inflammatory microenvironment. Our study gains a new mechanistic insight into how microglial Pd1 decides the fate of SCI and promotes microglial Pd1 as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Zhihao Lin
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China
| | - Xin Shen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Ming Li
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, 130033, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, 226001, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, 226001, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong Jiangsu Province, 226001, China.
| |
Collapse
|
17
|
Benavides FFW, Veldhuis Kroeze EJB, Leijten L, Schmitz KS, van Run P, Kuiken T, de Vries RD, Bauer L, van Riel D. Neuroinvasive and neurovirulent potential of SARS-CoV-2 in the acute and post-acute phase of intranasally inoculated ferrets. PLoS One 2025; 20:e0311449. [PMID: 40193353 PMCID: PMC11975070 DOI: 10.1371/journal.pone.0311449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/12/2025] [Indexed: 04/09/2025] Open
Abstract
Severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) can cause systemic disease, including neurological complications, even after mild respiratory disease. Previous studies have shown that SARS-CoV-2 infection can induce neurovirulence through microglial activation in the brains of patients and experimentally inoculated animals, which are models representative for moderate to severe respiratory disease. Here, we aimed to investigate the neuroinvasive and neurovirulent potential of SARS-CoV-2 in intranasally inoculated ferrets, a model for subclinical to mild respiratory disease. The presence of viral RNA, histological lesions, virus-infected cells, and the number and surface area of microglia and astrocytes were investigated. Viral RNA was detected in various respiratory tissue samples by qPCR at 7 days post inoculation (dpi). Virus antigen was detected in the nasal turbinates of ferrets sacrificed at 7 dpi and was associated with inflammation. Viral RNA was detected in the brains of ferrets sacrificed 7 dpi, but in situ hybridization nor immunohistochemistry did confirm evidence for viral RNA or antigen in the brain. Histopathological analysis of the brains showed no evidence for an influx of inflammatory cells. Despite this, we observed an increased number of Alzheimer type II astrocytes in the hindbrains of SARS-CoV-2 inoculated ferrets. Additionally, we detected increased microglial activation in the olfactory bulb and hippocampus, and a decrease in the astrocytic activation status in the white matter and hippocampus of SARS-CoV-2 inoculated ferrets. In conclusion, although SARS-CoV-2 has limited neuroinvasive potential in this model for subclinical to mild respiratory disease, there is evidence for neurovirulent potential. This study highlights the value of this ferret model to study the neuropathogenecity of SARS-CoV-2 and reveals that a mild SARS-CoV-2 infection can affect both microglia and astrocytes in different parts of the brain.
Collapse
Affiliation(s)
| | | | - Lonneke Leijten
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | | | - Peter van Run
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Thijs Kuiken
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Rory D. de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Lisa Bauer
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
18
|
Cui Y, Zhi SM, Ding PF, Zhu T, Chen XX, Liu XZ, Sheng B, Li XJ, Wang J, Zhang JT, Xu MX, Jiang YX, Hang CH, Li W. Silybin attenuates microglia-mediated neuroinflammation via inhibition of STING in experimental subarachnoid hemorrhage. Int Immunopharmacol 2025; 151:114305. [PMID: 39986195 DOI: 10.1016/j.intimp.2025.114305] [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: 08/31/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND The primary cause of subarachnoid hemorrhage (SAH) is the rupture of intracranial aneurysms. Over-activation of microglia following SAH is a primary driving force in early brain injury (EBI), which is a leading cause of poor outcomes. Silybin is a flavonoid compound extracted from Silybum marianum, a plant belonging to the Asteraceae family. Its anti-inflammatory and antioxidant properties could provide neuroprotective effects. The mechanism of silybin on EBI after SAH is unclear. PURPOSE To determine the therapeutic effect of silybin on SAH and its underlying mechanisms. METHODS We used a prechiasmatic autologous arterial blood injection in vivo and hemoglobin in vitro to establish experimental SAH model. Dexamethasone was used as a positive control drug. We evaluated the neuroprotective effect of silybin on the in vivo SAH model by neurological function scores, rotarod test, and open field test, and explored the protective effect of silybin on neuroinflammation and apoptosis after SAH by quantitative polymerase chain reaction (qPCR), western blot (WB), Immunofluorescence (IF) and TUNEL staining. IF staining of CD86 and CD206 was used to assess microglial phenotype polarization. Then we used WB and IF labeling of STING to explore the effect of silybin on the STING pathway after SAH, and used a combination of transcriptomics and non-targeted metabolomics to study the potential mechanism of silybin in detail, and verified the essential genes by qPCR. We also extracted cerebrospinal fluid from SAH patients and detected the expression level of STING in cerebrospinal fluid by enzyme-linked immunosorbent assay (ELISA) to clarify the association between STING and neural function. RESULTS Results showed that silybin ameliorated neuronal damage and improved short-term neurological function, and reduced inflammatory damage and neuronal apoptosis in SAH mice. Silybin inhibited the expression levels of TNF-α, IL-1β and NLRP3, and promoted the expression levels of CD206, Arg1 and IL-10. Notably, Silybin promoted M2 microglia polarization. Further studies found that silybin reduced the mRNA and protein levels of the stimulator of interferon genes (STING) in microglia. And the use of a specific activator of STING (CMA) disrupted the protective effect of silybin. A total of 358 differential expression genes were identified using transcriptomics, and 150 different metabolites abundance were identified using metabolomic screening. Analysis of the effects of STING on transcriptomics and metabolomics revealed that STING might impact metabolic pathways, including linoleic acid metabolism. The qPCR results confirmed the decreased expression of essential proteins involved in the pathway. Finally, we found that increased STING expression in the cerebrospinal fluid of SAH patients was associated with decreased neurological function scores and poor prognosis. CONCLUSION Silybin had a therapeutic effect on SAH. The underlying mechanism involves linoleic acid metabolism, which is associated with the differential genes and metabolites detected in the study. This study presented a pharmacological rationale for using silybin to treat SAH.
Collapse
Affiliation(s)
- Yue Cui
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Si-Min Zhi
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Peng-Fei Ding
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Ting Zhu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Xiang-Xin Chen
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xun-Zhi Liu
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bin Sheng
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiao-Jian Li
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Wang
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jia-Tong Zhang
- Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meng-Xiao Xu
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Yong-Xiao Jiang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Neurosurgical Institute, Nanjing University; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
19
|
Kaur A, Aran KR. Unraveling the cGAS-STING pathway in Alzheimer's disease: A new Frontier in neuroinflammation and therapeutic strategies. Neuroscience 2025; 573:430-441. [PMID: 40185388 DOI: 10.1016/j.neuroscience.2025.04.001] [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: 01/07/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent type of neurological disorder characterized by cognitive decline and memory loss, marked by the accumulation of amyloid beta (Aβ) plaques and hyperphosphorylated tau protein, causing extensive neuronal death and neuroinflammation. There is growing evidence that AD development extends beyond the neuronal compartment and has a major impact on the immunological functions of the brain. The cyclic GMP-AMP synthase (cGAS) detects cytosolic DNA, including pathogenic foreign DNA and self-DNA from cellular injury, triggering a type I interferon (IFN-I) response through activation of the stimulator of interferon genes (STING). The activation of the cGAS-STING pathway in response to mitochondrial dysfunction drives neuroinflammation in AD, which is mediated by the release of IFN-I cytokines. Furthermore, the release of oxidized mtDNA is necessary for the stimulation of the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome, which is a family of protein complexes that macrophages can produce to induce inflammation. AD becomes severe by the stimulation of the cGAS-STING pathway, which results in sterile inflammation and microglial dysfunction. This review aims to explore the potential impact of the cGAS-STING signaling pathway in the pathogenesis and progression of AD. Additionally; after overviewing recent findings, this article highlights the molecular mechanism involved in the onset of disease and its modulation regarding the therapeutic approach of AD. Finally, deliberated a deep insight, the cGAS-STING axis could provide novel therapeutic avenues for slowing or halting the progression of AD, thereby offering new prospects for treatment development.
Collapse
Affiliation(s)
- Arshdeep Kaur
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab 142001, India.
| |
Collapse
|
20
|
Luo W, Xu C, Li L, Ji Y, Wang Y, Li Y, Ye Y. Perfluoropentane-based oxygen-loaded nanodroplets reduce microglial activation through metabolic reprogramming. Neural Regen Res 2025; 20:1178-1191. [PMID: 38989955 PMCID: PMC11438333 DOI: 10.4103/nrr.nrr-d-23-01299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00032/figure1/v/2024-07-06T104127Z/r/image-tiff Microglia, the primary immune cells within the brain, have gained recognition as a promising therapeutic target for managing neurodegenerative diseases within the central nervous system, including Parkinson's disease. Nanoscale perfluorocarbon droplets have been reported to not only possess a high oxygen-carrying capacity, but also exhibit remarkable anti-inflammatory properties. However, the role of perfluoropentane in microglia-mediated central inflammatory reactions remains poorly understood. In this study, we developed perfluoropentane-based oxygen-loaded nanodroplets (PFP-OLNDs) and found that pretreatment with these droplets suppressed the lipopolysaccharide-induced activation of M1-type microglia in vitro and in vivo, and suppressed microglial activation in a mouse model of Parkinson's disease. Microglial suppression led to a reduction in the inflammatory response, oxidative stress, and cell migration capacity in vitro. Consequently, the neurotoxic effects were mitigated, which alleviated neuronal degeneration. Additionally, ultrahigh-performance liquid chromatography-tandem mass spectrometry showed that the anti-inflammatory effects of PFP-OLNDs mainly resulted from the modulation of microglial metabolic reprogramming. We further showed that PFP-OLNDs regulated microglial metabolic reprogramming through the AKT-mTOR-HIF-1α pathway. Collectively, our findings suggest that the novel PFP-OLNDs constructed in this study alleviate microglia-mediated central inflammatory reactions through metabolic reprogramming.
Collapse
Affiliation(s)
- Wanxian Luo
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chuanhui Xu
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Linxi Li
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yunxiang Ji
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yingjia Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yongyi Ye
- Institute of Neuroscience, Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
21
|
Wu J, Xu W, Su Y, Wang GH, Ma JJ. Targeting chaperone-mediated autophagy in neurodegenerative diseases: mechanisms and therapeutic potential. Acta Pharmacol Sin 2025; 46:816-828. [PMID: 39548290 PMCID: PMC11950187 DOI: 10.1038/s41401-024-01416-3] [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: 07/03/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
The pathological hallmarks of various neurodegenerative diseases including Parkinson's disease and Alzheimer's disease prominently feature the accumulation of misfolded proteins and neuroinflammation. Chaperone-mediated autophagy (CMA) has emerged as a distinct autophagic process that coordinates the lysosomal degradation of specific proteins bearing the pentapeptide motif Lys-Phe-Glu-Arg-Gln (KFERQ), a recognition target for the cytosolic chaperone HSC70. Beyond its role in protein quality control, recent research underscores the intimate interplay between CMA and immune regulation in neurodegeneration. In this review, we illuminate the molecular mechanisms and regulatory pathways governing CMA. We further discuss the potential roles of CMA in maintaining neuronal proteostasis and modulating neuroinflammation mediated by glial cells. Finally, we summarize the recent advancements in CMA modulators, emphasizing the significance of activating CMA for the therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin Wu
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| | - Wan Xu
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Ying Su
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Guang-Hui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Jing-Jing Ma
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
22
|
Xu W, Huang Y, Zhou R. NLRP3 inflammasome in neuroinflammation and central nervous system diseases. Cell Mol Immunol 2025; 22:341-355. [PMID: 40075143 PMCID: PMC11955557 DOI: 10.1038/s41423-025-01275-w] [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: 12/06/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of various central nervous system (CNS) diseases. The NLRP3 inflammasome is an important intracellular multiprotein complex composed of the innate immune receptor NLRP3, the adaptor protein ASC, and the protease caspase-1. The activation of the NLRP3 inflammasome can induce pyroptosis and the release of the proinflammatory cytokines IL-1β and IL-18, thus playing a central role in immune and inflammatory responses. Recent studies have revealed that the NLRP3 inflammasome is activated in the brain to induce neuroinflammation, leading to further neuronal damage and functional impairment, and contributes to the pathological process of various neurological diseases, such as multiple sclerosis, Parkinson's disease, Alzheimer's disease, and stroke. In this review, we summarize the important role of the NLRP3 inflammasome in the pathogenesis of neuroinflammation and the pathological course of CNS diseases and discuss potential approaches to target the NLRP3 inflammasome for the treatment of CNS diseases.
Collapse
Grants
- 81821001, 82130107, 82330052, 82202038, U20A20359 National Natural Science Foundation of China (National Science Foundation of China)
- National Key research and development program of China (grant number (2020YFA0509101), The Strategic Priority Research Program of the Chinese Academy of Sciences (XDB0940000),
- MEXT | JST | Strategic Promotion of Innovative R and D (Strategic Promotion of Innovative R&D)
- the CAS Project for Young Scientists in Basic Research (YSBR-074) and the Fundamental Research Funds for the Central Universities, the outstanding Youth Project of Anhui Provincial Natural Science Foundation (2408085Y049), the Research Start-up Funding of the Institute of Health and Medicine, Hefei Comprehensive National Science Center (2024KYQD004), the Natural Science Foundation of Jiangsu Province (BK20221085),
- The key project of Anhui Provincial Department of Education Fund (2024AH052060).
Collapse
Affiliation(s)
- Wen Xu
- Neurology Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Yi Huang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
| | - Rongbin Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
23
|
Cao LH, Wang ZZ, Zhao H, Tian S, He HJ, Miao JX, Huang SN, Wang XY, Song YG, Kang L, Wang HY, Wang C, Bai M, Gao S, Miao MS. The microglial state transition as a novel mechanism by which fresh Baihe Dihuang decoction prevents depression by regulating SIRT1/HMGB1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156718. [PMID: 40220404 DOI: 10.1016/j.phymed.2025.156718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Fresh Baihe Dihuang decoction (FBD) is a classic Traditional Chinese Medicine (TCM) formula used to treat depression. However, its underlying molecular mechanisms in treating depression still need further exploration. PURPOSE In this study, we investigated whether FBD could prevent depressive-like behaviors by remodeling the microglial state transition via the inhibition of SIRT1/HMGB1 signaling in vivo and in vitro. STUDY DESIGN AND METHODS In vivo, adult male C57BL/6 J mice were subjected to chronic unpredictable mild stress (CUMS) for 6 weeks to investigate whether FBD could produce antidepressant-like behavioral effects. At 9 weeks of age, EX527, a SIRT1 inhibitor, was injected intraperitoneally 30 min before the intragastric administration of FBD, Flx or vehicle daily until 12 weeks of age, and the effects of alterations in SIRT1/HMGB1 signaling on CUMS-mediated depression were investigated. In vitro, the anti-depressive mechanism of FBD was further investigated using BV-2 cells (a microglial cell line) and primary PFC neurons. We examined depression-like behaviors using behavioral tests. Serum and supernatants samples were collected and interleukin-1β (IL-1β), IL-6 and tumor necrosis factor-α (TNF-α) levels were measured using enzyme-linked immunosorbent assays (ELISAs). Prefrontal cortical (PFC) tissues, BV-2 cells and PFC neurons were collected to detect neuronal apoptosis, the microglial state or proteins in the silent information regulator 2 homolog 1 (SIRT1)/high mobility group box 1 (HMGB1) signaling pathway via flow cytometry, immunohistochemical staining, immunofluorescence staining,TUNEL staining or western blot analysis. RESULTS The administration of FBD ameliorated the depressive-like behaviors induced by CUMS. In addition, FBD supplementation promoted the transition from a proinflammatory microglial phenotype to an anti-inflammatory microglial phenotype. The FBD-mediated inhibition of HMGB1 expression and its nucleocytoplasmic translocation were identified as likely due to increased SIRT1 activity, effectively inhibiting the subsequent inflammatory response. Furthermore, our findings revealed that FBD notably attenuated neuronal apoptosis in the PFC. In contrast, the SIRT1 inhibitor EX527 counteracted the antidepressant effect of FBD while also reversing the expression of brain-derived neurotrophic factor (BDNF), NeuN, cleaved caspase-3, bax, and bcl-2 proteins. CONCLUSIONS This study showed that FBD prevents depression by mediating a microglial state transition via the SIRT1/HMGB1 signaling pathway, providing a promising and novel antidepressant therapeutic strategy.
Collapse
Affiliation(s)
- Li-Hua Cao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Zhen-Zhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hui Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Shuo Tian
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hong-Juan He
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jin-Xin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Sheng-Nan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Xiao-Yu Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Ya-Gang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Le Kang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Hao-Yang Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Can Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Ming Bai
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Song Gao
- Henan Solid Formulation Technology Innovation Center, Zhongjing Wanxi Pharmaceutical Co., Ltd., Xixia 474550, Henan Province, China
| | - Ming-San Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China.
| |
Collapse
|
24
|
Liu C, Zhao D, Yu G, Du H, Xu L, Cao Y, Cui M, Wang W, Wang D, Liu J, Meng F, Hu F, Li W, Du J, Li C. Alleviation of Microglia Mediating Hippocampal Neuron Impairments and Depression-Related Behaviors by Urolithin B via the SIRT1-FOXO1 Pathway. CNS Neurosci Ther 2025; 31:e70379. [PMID: 40237232 DOI: 10.1111/cns.70379] [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: 10/09/2024] [Revised: 03/17/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025] Open
Abstract
AIMS Conventional antidepressants exhibit limited efficacy and delayed onset. This study aimed to elucidate the antidepressant effects of urolithin B (UB) and its regulatory role in microglia-mediated hippocampal neuronal dysfunction. METHODS The mouse model of depression was established using both chronic unpredicted stress (CUS) and lipopolysaccharide (LPS) injection. The therapeutic efficacy of UB was assessed through behavioral paradigms. The microglia activation, cellular cytotoxicity and apoptosis levels, and underlying molecular mechanisms were delineated utilizing proteomics analysis, immunofluorescence staining, real-time PCR and Western blotting. RESULTS UB efficiently alleviated depression-related behaviors, accompanied by suppressed microglia activation, neuroinflammation, changes of classic activation (M1)/alternative activation (M2) polarization and recovered sirtuin-1 (SIRT1) and forkhead box protein O1 (FOXO1) expression in the hippocampus. Additionally, UB reduced the cytotoxicity and apoptosis of HT22 cells and depression-related phenotypes treated by the cellular supernatant from LPS-incubated BV2 cells, which was mediated by the SIRT1-FOXO1 pathway. The proteomics analysis of the cellular supernatant content revealed abundant secreting proteins among the LPS/UB application. CONCLUSION This study confirmed that microglial SIRT1 mediates UB's antidepressant effects, positioning UB as a promising therapeutic candidate for depression by targeting neuroinflammatory pathways.
Collapse
Affiliation(s)
- Cuilan Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Di Zhao
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Guoxing Yu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - HengWei Du
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yifan Cao
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Minghu Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wentao Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fengai Hu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wei Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Du
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Rehabilitation Medicine, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
25
|
Fayyad-kazan M, Soayfane Z, Faour W, Fayyad-Kazan H, Awada R. Autotaxin regulates the expression and the activity of P-glycoprotein in lipopolysaccharide -activated microglial cells. Cytotechnology 2025; 77:58. [PMID: 39959786 PMCID: PMC11822174 DOI: 10.1007/s10616-025-00727-5] [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: 11/13/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer's and Parkinson's, are characterized by chronic inflammation and oxidative stress, often mediated by activated microglial cells. Microglia-induced neuroinflammation is essential to neuronal damage, driven by the overproduction of pro-inflammatory cytokines and reactive oxygen species. Autotaxin (ATX), a lysophospholipase D enzyme, can modulate inflammation through its enzymatic product lysophosphatidic acid (LPA). While previous studies highlighted ATX's anti-inflammatory properties, its impact on P-glycoprotein (P-gp), a key efflux transporter involved in drug resistance and neuroinflammation, remains not fully understood. The objective of this study was to explore how ATX modulates the expression and activity of P-gp in lipopolysaccharide (LPS)-activated and H2O2-stressed BV-2 microglial cells. Microglial cells were transfected with either an empty vector (EV) or an ATX cDNA vector (A +) and exposed to LPS (1 µg/mL) or H2O2 (100 µM). The mRNA expression levels of P-gp and pro-inflammatory cytokines were analyzed using qRT-PCR, and P-gp activity was assessed using the NBD-CSA fluorescence efflux assay. Our findings revealed that while LPS- and H2O2-treated microglial cells were characterized by an abnormal cellular morphology with long ramified processes, ATX overexpression restored the round shape morphology normally observed in the control untreated cells. Interestingly, ATX overexpression significantly reduced the mRNA levels of pro-inflammatory cytokines, such as TNF-α, in LPS- and H2O2-treated microglial cells. Moreover, ATX overexpression reduced both the mRNA levels and efflux activity of P-gp under inflammatory and oxidative stress conditions. These results suggest that ATX mitigates microglial activation and its downstream effects, highlighting its therapeutic potential in reducing neuroinflammation.
Collapse
Affiliation(s)
- Mohammad Fayyad-kazan
- Department of Natural and Applied Sciences, School of Arts and Sciences, The American University of Iraq – Baghdad, Baghdad, Iraq
| | - Zeina Soayfane
- Anticancer Therapeutic Approaches Group (ATAC), Rammal Rammal Laboratory, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Wissam Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Hussein Fayyad-Kazan
- Anticancer Therapeutic Approaches Group (ATAC), Rammal Rammal Laboratory, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Rana Awada
- Anticancer Therapeutic Approaches Group (ATAC), Rammal Rammal Laboratory, Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
26
|
Liu Y, Cai X, Shi B, Mo Y, Zhang J, Luo W, Yu B, Li X. Mechanisms and Therapeutic Prospects of Microglia-Astrocyte Interactions in Neuropathic Pain Following Spinal Cord Injury. Mol Neurobiol 2025; 62:4654-4676. [PMID: 39470872 DOI: 10.1007/s12035-024-04562-1] [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: 05/24/2024] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
Neuropathic pain is a prevalent and debilitating condition experienced by the majority of individuals with spinal cord injury (SCI). The complex pathophysiology of neuropathic pain, involving continuous activation of microglia and astrocytes, reactive gliosis, and altered neuronal plasticity, poses significant challenges for effective treatment. This review focuses on the pivotal roles of microglia and astrocytes, the two major glial cell types in the central nervous system, in the development and maintenance of neuropathic pain after SCI. We highlight the extensive bidirectional interactions between these cells, mediated by the release of inflammatory mediators, neurotransmitters, and neurotrophic factors, which contribute to the amplification of pain signaling. Understanding the microglia-astrocyte crosstalk and its impact on neuronal function is crucial for developing novel therapeutic strategies targeting neuropathic pain. In addition, this review discusses the fundamental biology, post-injury pain roles, and therapeutic prospects of microglia and astrocytes in neuropathic pain after SCI and elucidates the specific signaling pathways involved. We also speculated that the extracellular matrix (ECM) can affect the glial cells as well. Furthermore, we also mentioned potential targeted therapies, challenges, and progress in clinical trials, as well as new biomarkers and therapeutic targets. Finally, other relevant cell interactions in neuropathic pain and the role of glial cells in other neuropathic pain conditions have been discussed. This review serves as a comprehensive resource for further investigations into the microglia-astrocyte interaction and the detailed mechanisms of neuropathic pain after SCI, with the aim of improving therapeutic efficacy.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xintong Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yajie Mo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianmin Zhang
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenting Luo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
27
|
Shi Q, Gutierrez RA, Bhat MA. Microglia, Trem2, and Neurodegeneration. Neuroscientist 2025; 31:159-176. [PMID: 38769824 PMCID: PMC11576490 DOI: 10.1177/10738584241254118] [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: 05/22/2024]
Abstract
Microglia are a specialized type of neuroimmune cells that undergo morphological and molecular changes through multiple signaling pathways in response to pathological protein aggregates, neuronal death, tissue injury, or infections. Microglia express Trem2, which serves as a receptor for a multitude of ligands enhancing their phagocytic activity. Trem2 has emerged as a critical modulator of microglial activity, especially in many neurodegenerative disorders. Human TREM2 mutations are associated with an increased risk of developing Alzheimer disease (AD) and other neurodegenerative diseases. Trem2 plays dual roles in neuroinflammation and more specifically in disease-associated microglia. Most recent developments on the molecular mechanisms of Trem2, emphasizing its role in uptake and clearance of amyloid β (Aβ) aggregates and other tissue debris to help protect and preserve the brain, are encouraging. Although Trem2 normally stimulates defense mechanisms, its dysregulation can intensify inflammation, which poses major therapeutic challenges. Recent therapeutic approaches targeting Trem2 via agonistic antibodies and gene therapy methodologies present possible avenues for reducing the burden of neurodegenerative diseases. This review highlights the promise of Trem2 as a therapeutic target, especially for Aβ-associated AD, and calls for more mechanistic investigations to understand the context-specific role of microglial Trem2 in developing effective therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Qian Shi
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Raul A. Gutierrez
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, Center for Biomedical Neuroscience, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| |
Collapse
|
28
|
Zhang Y, Tang Y, Illes P. Modification of Neural Circuit Functions by Microglial P2Y6 Receptors in Health and Neurodegeneration. Mol Neurobiol 2025; 62:4139-4148. [PMID: 39400857 PMCID: PMC11880064 DOI: 10.1007/s12035-024-04531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Neural circuits consisting of neurons and glial cells help to establish all functions of the CNS. Microglia, the resident immunocytes of the CNS, are endowed with UDP-sensitive P2Y6 receptors (P2Y6Rs) which regulate phagocytosis/pruning of excessive synapses during individual development and refine synapses in an activity-dependent manner during adulthood. In addition, this type of receptor plays a decisive role in primary (Alzheimer's disease, Parkinson's disease, neuropathic pain) and secondary (epilepsy, ischemic-, mechanical-, or irradiation-induced) neurodegeneration. A whole range of microglial cytokines controlled by P2Y6Rs, such as the interleukins IL-1β, IL-6, IL-8, and tumor necrosis factor-α (TNF-α), leads to neuroinflammation, resulting in neurodegeneration. Hence, small molecular antagonists of P2Y6Rs and genetic knockdown of this receptor provide feasible ways to alleviate inflammation-induced neurological disorders but might also interfere with the regulation of the synaptic circuitry. The present review aims at investigating this dual role of P2Y6Rs in microglia, both in shaping neural circuits by targeted phagocytosis and promoting neurodegenerative illnesses by fostering neuroinflammation through multiple transduction mechanisms.
Collapse
Affiliation(s)
- Yi Zhang
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Peter Illes
- International Joint Research Centre on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
29
|
Li L, Gao W, Ren N, Chen L. IL-2/anti-IL-2 complexes attenuates neuroinflammation and neurodegeneration in mice of experimental Parkinson's disease. Brain Res Bull 2025; 223:111273. [PMID: 39999936 DOI: 10.1016/j.brainresbull.2025.111273] [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: 10/14/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 02/27/2025]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, with motor and non-motor symptoms being its main clinical manifestations. Neuroinflammation has been shown to involve in pathogenesis of PD. Regulatory T cells (Tregs) in PD exhibited reduction in number and suppressive activity. Existing methods to increase the Tregs remains challenging for clinical application because of the difficulty in Tregs expanding or serious side-effects. Therefore, new approaches still need to be explored to balance the amount and activity of Tregs. In this study, we assessed the protective effects of IL-2/anti-IL-2 complexes (IL-2C) on mouse models of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). And the results showed that IL-2C significantly increased the number of Tregs both in spleen and brain, accompanied by reduced nigral dopaminergic neuron loss and behavioral defects. Besides, IL-2C also attenuated neuroinflammation as observed by diminished glial activation, fewer infiltration of CD4+ and CD8+ T cells and reduced pro-inflammatory cytokines releasing in the nigral region. Moreover, the protective effects of IL-2C were abolished by pre-treatment of anti-CD25 antibody (PC61), which was used to delete the Tregs. In summary, our results demonstrate that IL-2C-induced Tregs expansion attenuates the dopaminergic neurons loss and the neuroinflammatory response in vivo, suggesting that IL-2C maybe a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Lanxin Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China
| | - Weiwei Gao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China.
| | - Ning Ren
- Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China
| | - Lei Chen
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Department of Neurology, Tianjin Huanhu Hospital, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin 300350, China; Tianjin Neurosurgical Institute, NO. 6 Jizhao Road, Jinnan District, Tianjin 300350, China.
| |
Collapse
|
30
|
Stéen EJL, Tousiaki EN, Kingston L, van der Wildt B, Lénárt N, Beaino W, Verlaan M, Zarzycka B, Zinnhardt B, Dénes Á, Gobbi L, de Esch IJP, Elmore CS, Windhorst AD, Honer M, Leurs R. Characterization of [ 3H]AZ12464237 as a high affinity, non-nucleotide antagonist radioligand for the P2Y 12 receptor. Biochem Pharmacol 2025; 237:116900. [PMID: 40179990 DOI: 10.1016/j.bcp.2025.116900] [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: 11/11/2024] [Revised: 02/26/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
The purinergic receptor P2Y12 (P2Y12R) is a well-recognized target for anti-thrombotic agents. This receptor is also expressed in microglia, the main immune cells of the brain, where it modulates microglial activation states and inflammatory responses. To investigate P2Y12R-mediated actions in the central nervous system (CNS), developing novel brain-penetrant ligands and further in vitro studies on brain tissues are essential. A radiolabeled, easily accessible tool compound would significantly advance such drug discovery efforts. Herein, we describe the 3H-labeling of a non-nucleotide P2Y12R antagonist AZ12464237, and its in vitro binding properties to the receptor in membrane preparations from transfected cells, as well as on mouse brain tissues. The radioligand shows high affinity toward both the human and rat P2Y12R, with Kd values of 3.12 ± 0.70 nM (human) and 16.6 ± 3.40 nM (rat), as determined by saturation binding studies. The binding kinetics of [3H]AZ12464237 are rapid with a short target residence time (∼1 min). We further confirmed the selectivity of the radioligand by performing competitive displacement studies, in which reported P2Y12R ligands and other P2Y receptors ligands were tested for binding against [3H]AZ12464237. Additionally, the radioligand proved valuable for in vitro autoradiography studies on mouse brain tissues, although limited off-target binding was observed in P2Y12R knock-out mouse brain. This could be traced to glycogen synthase kinase 3 α. Considering the growing interest in P2Y12R as a biomarker for anti-inflammatory microglia, [3H]AZ12464237 represents a promising tool for in vitro studies, including screening assays aimed at identifying novel P2Y12R ligands for CNS applications.
Collapse
Affiliation(s)
- E Johanna L Stéen
- Amsterdam Neuroscience, Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute of Molecules, Medicines and Systems, Department of Medicinal Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| | - Efthalia-Natalia Tousiaki
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Neuroscience and Rare Diseases, Biomarkers, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Lee Kingston
- Isotope Chemistry, Early Chemical Development, Pharmaceutical Sciences R&D, AstraZeneca, Gothenburg, Sweden
| | - Berend van der Wildt
- Amsterdam Neuroscience, Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Nikolett Lénárt
- "Momentum", Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Wissam Beaino
- Amsterdam Neuroscience, Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mariska Verlaan
- Amsterdam Neuroscience, Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Barbara Zarzycka
- Amsterdam Institute of Molecules, Medicines and Systems, Department of Medicinal Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Bastian Zinnhardt
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Neuroscience and Rare Diseases, Biomarkers, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ádám Dénes
- "Momentum", Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Luca Gobbi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Therapeutic Modalities, Small Molecule Research, Medicinal Chemistry, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Iwan J P de Esch
- Amsterdam Institute of Molecules, Medicines and Systems, Department of Medicinal Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Charles S Elmore
- Isotope Chemistry, Early Chemical Development, Pharmaceutical Sciences R&D, AstraZeneca, Gothenburg, Sweden
| | - Albert D Windhorst
- Amsterdam Neuroscience, Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Michael Honer
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Neuroscience and Rare Diseases, Biomarkers, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Rob Leurs
- Amsterdam Institute of Molecules, Medicines and Systems, Department of Medicinal Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
31
|
Chen Y, Yao X, Wang C, Zhuang H, Xie B, Sun C, Wang Z, Zhou X, Luo Y, Zhang Y, Zhou S, Liu L. Minocycline treatment attenuates neurobehavioural abnormalities and neurostructural aberrations in the medial prefrontal cortex in mice fed a high-fat diet during adolescence. Brain Behav Immun 2025; 128:83-98. [PMID: 40180016 DOI: 10.1016/j.bbi.2025.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025] Open
Abstract
A preference for and overconsumption of a high-fat diet (HFD) are common among adolescents and are recognized as risk factors for multiple mental disorders. The protracted maturation of the medial prefrontal cortex (mPFC), a key brain structure that plays a critical role in mental functions that are essential for both developing and mature individuals (including emotional processing, decision making, risk assessment, and creative thinking), during adolescence renders it more vulnerable to the environmental insults experienced during this critical developmental window. However, the effects of HFD consumption during adolescence on mPFC-related behaviours and the underlying mechanisms need to be further investigated. In this study, we observed that mice fed a HFD throughout adolescence developed depressive- and anxiety-like behaviours and distinctively increased risk-avoidance behaviour, accompanied by morphological aberrations of both pyramidal neuron and microglia in the mPFC. The systemic administration of minocycline, a well-known broad-spectrum antibiotic, effectively attenuated the adverse effects of HFD consumption during adolescence on neurobehaviours and the morphology of pyramidal neurons in the mPFC. This study provides new insights into the psychological effects of long-term HFD consumption during adolescence and indicates the existence of a window during which microglial stabilization may be a promising strategy to protect against the HFD consumption-induced increase in the risk of psychiatric disorders.
Collapse
Affiliation(s)
- Yuxi Chen
- Medical College, Southeast University, Nanjing 210009, China
| | - Xiuting Yao
- Medical College, Southeast University, Nanjing 210009, China
| | - Conghui Wang
- Medical College, Southeast University, Nanjing 210009, China
| | - Hong Zhuang
- Medical College, Southeast University, Nanjing 210009, China
| | - Bingjie Xie
- Medical College, Southeast University, Nanjing 210009, China
| | - Congli Sun
- Medical College, Southeast University, Nanjing 210009, China
| | - Zixuan Wang
- Medical College, Southeast University, Nanjing 210009, China
| | - Xinguo Zhou
- Medical College, Southeast University, Nanjing 210009, China
| | - Yu Luo
- Medical College, Southeast University, Nanjing 210009, China
| | - Yilin Zhang
- Medical College, Southeast University, Nanjing 210009, China
| | - Shihui Zhou
- Medical College, Southeast University, Nanjing 210009, China
| | - Lijie Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
32
|
ElNebrisi E, Lozon Y, Oz M. The Role of α7-Nicotinic Acetylcholine Receptors in the Pathophysiology and Treatment of Parkinson's Disease. Int J Mol Sci 2025; 26:3210. [PMID: 40244021 DOI: 10.3390/ijms26073210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
The α7 nicotinic acetylcholine receptor (α7-nAChR) is a pivotal regulator of neurotransmission, neuroprotection, and immune modulation in the central nervous system. This review explores its structural and functional attributes, highlighting its therapeutic potential in neurodegenerative disorders, particularly Parkinson's disease (PD). α7-nAChRs mediate synaptic plasticity, modulate inflammatory responses, and influence dopamine release, positioning them as a promising pharmacological target. Positive allosteric modulators (PAMs) enhance α7-nAChR activity mainly by reducing desensitization, offering a superior therapeutic approach compared with direct agonists. Emerging preclinical studies suggest that α7-nAChR activation mitigates dopaminergic neurodegeneration, improves L-dopa-induced dyskinesia, and reduces neuroinflammation. Despite promising findings, clinical trials have yielded mixed results, necessitating further research into optimizing α7-targeted therapies. This review underscores the significance of α7-nAChRs in PD pathophysiology and highlights future directions for their translational potential in neuroprotection and symptomatic relief.
Collapse
Affiliation(s)
- Eslam ElNebrisi
- Department of Biomedical Sciences, Dubai Medical College for Girls, Dubai Medical University, Dubai 20170, United Arab Emirates
| | - Yosra Lozon
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai Medical University, Dubai 20170, United Arab Emirates
| | - Murat Oz
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Safat 13110, Kuwait
| |
Collapse
|
33
|
Zhao X, Li D, Zhu Z, Li S, Qin Y, Yang Y. Spermidine attenuates microglial activation, neuroinflammation, and neuronal injury in rat model of vascular dementia. Neuroscience 2025; 573:355-363. [PMID: 40169099 DOI: 10.1016/j.neuroscience.2025.03.054] [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: 05/29/2024] [Revised: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 04/03/2025]
Abstract
Spermidine has been implicated to provide beneficial effects on cognitive function in several model organisms as well as older adults with mild and moderate dementia. Nevertheless, the potential impact of spermidine on learning and memory deficits in vascular dementia (VaD) remains largely unknown. Here, bilateral common carotid artery occlusion (BCCAo) was applied to induce chronic cerebral hypoperfusion in rats. We demonstrated that spermidine therapy improved the spatial learning performance in model animals, accompanied with decreased cerebral histopathologic injury and increased restored myelin basic protein (MBP) expression. Moreover, spermidine suppressed abnormal microglia activation, inhibited the excessive generation of proinflammatory mediators, such as tumor necrosis factor (TNF)α and inducible nitric oxide synthase (iNOS), and increased the anti-inflammatory cytokine transforming growth factor (TGF)β expression in rodent brain following hypoperfusion. Our findings indicated that spermidine alleviated cognitive impairments of rats after VaD-like injury possibly via suppressing microglia-modulated neuroinflammation and neuronal injury. These data may shed light on understanding the pathogenesis of VaD and point to the promising value of spermidine supplementation for cognition improvement.
Collapse
Affiliation(s)
- Xinyuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Dan Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Zirui Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Suzhen Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Yaze Qin
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Yi Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
34
|
Kim YS, Lee SB, Kang SI, Kim WJ, Choi DK. A Chloroform Fraction Derived from Vitis vinifera Root Ethanol Extract Attenuates Lipopolysaccharide-Induced Inflammatory Responses and Cognitive Dysfunction in BV-2 Microglia Cells and C57BL/6J Mouse Model. Int J Mol Sci 2025; 26:3126. [PMID: 40243887 DOI: 10.3390/ijms26073126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
This study aimed to investigate the inhibitory effect of the chloroform fraction (CF) from Vitis vinifera root extract on LPS-induced neuroinflammation in BV-2 microglia cells and a C57/BL6J mouse model. CF significantly suppressed LPS-induced proinflammatory cytokines, including nitric oxide (NO), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in BV-2 microglia cells. Mechanistically, CF inhibited LPS-induced activation of nuclear factor-κB (NF-κB) by blocking the p65 subunit and preventing the phosphorylation of NF-kappa-B inhibitor α (IκBα), while its effect was independent of the mitogen-activated protein kinase (MAPK) pathway. Furthermore, CF modulated the TRIF signaling pathway by regulating TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3), which contributed to the suppression of inflammatory mediators in BV-2 microglia cells. In vivo, we evaluated the neuroprotective effects of CF against cognitive dysfunction and inflammatory responses in an LPS-induced mouse model. Our behavioral assessments, including the Morris water maze and Y-maze tests, demonstrated that CF alleviated LPS-induced spatial learning impairment and cognitive decline. Additionally, CF significantly reduced the levels of inflammatory cytokines in serum and inflammatory mediators proteins expression in whole brain in LPS-injected mice, suggesting a direct link between reduced inflammatory responses and improved cognitive function. These findings suggest that CF from V. vinifera root extract may serve as a potential therapeutic strategy for neurodegenerative diseases mediated by microglial activation, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Yon-Suk Kim
- Department of Biotechnology, Research Institute of Inflammatory Disease (RID), College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Sang-Bong Lee
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Shin-Il Kang
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Woo-Jung Kim
- Biocenter, Gyeonggido Business and Science Accelerator, Gwanggyo-ro 147, Yeongtong-gu, Suwon 16229, Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, Research Institute of Inflammatory Disease (RID), College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
- Department of Applied Life Sciences, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
35
|
Yagi H, Boeck M, Neilsen K, Yang J, Ko M, Tomita Y, Negishi K, Fu Z, Sun Y, Smith LEH. Choroidal Neovascularization Is Suppressed With Activation of TREM2 in Mononuclear Phagocytes. Arterioscler Thromb Vasc Biol 2025. [PMID: 40143815 DOI: 10.1161/atvbaha.124.321809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND Mononuclear phagocytes contribute to pathological angiogenesis in age-related macular degeneration, a leading worldwide cause of visual impairment. However, the mechanisms that orchestrate the functions of mononuclear phagocytes remain poorly understood. TREM2 (triggering receptor on myeloid cells 2) has been shown to be crucial for the activation of mononuclear phagocytes in atherosclerosis, fatty liver disease, and Alzheimer disease. The objective of this study was to investigate the role of TREM2 in pathological angiogenesis in age-related macular degeneration. METHODS C57BL/6J and Trem2 knockout mice were subjected to laser-induced choroidal neovascularization, a model of choroidal neovascular age-related macular degeneration. Purified bovine sulfatide and agonist anti-TREM2 antibody was used to activate TREM2 signaling. The expression of TREM2 or downstream signals were assessed with immunohistochemistry or qPCR. In vitro murine macrophage RAW264.7 cells were used to investigate the direct impact of sulfatide on inflammatory and phagocytic responses. RESULTS We found that pharmacological activation of TREM2 suppressed laser-induced choroidal neovessel formation. The activation of TREM2 in mononuclear phagocytes suppressed TNF (tumor necrosis factor) and subsequently promoted phagocytosis. CONCLUSIONS These findings demonstrate that activation of TREM2 in mononuclear phagocytes suppresses the proinflammatory response, promotes phagocytosis, and impedes choroidal neovessel formation. Our study provides insight into the critical role of TREM2 in pathological angiogenesis.
Collapse
Affiliation(s)
- Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan (H.Y., Y.T., K. Negishi)
| | - Myriam Boeck
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Germany (M.B.)
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| | - Jay Yang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| | - Minji Ko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan (H.Y., Y.T., K. Negishi)
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan (H.Y., Y.T., K. Negishi)
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, MA (H.Y., M.B., K. Neilsen, J.Y., M.K., Y.T., Z.F., Y.S., L.E.H.S.)
| |
Collapse
|
36
|
Hu W, Chen M, Lin Y, Zhang H, Sun L, Shao W, Ye Y, Cheng Y, Zhou S, Hu P, Wu X, Xu Y, Wang K. Neuronal CD47 induces behavioral alterations and ameliorates microglial synaptic pruning in wild-type and Alzheimer's mouse models. Cell Biosci 2025; 15:38. [PMID: 40140948 PMCID: PMC11948738 DOI: 10.1186/s13578-025-01378-x] [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: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Microglia are brain-resident macrophages that play a crucial role in synapse pruning during the development and progression of various neuropsychiatric disorders, including autism spectrum disorder (ASD) and Alzheimer's disease (AD). Mechanistically, CD47 protein acts as a potent 'do not eat me' signal, protecting synapses from phagocytosis by microglia. However, the functional role of the upregulated neuronal CD47 signal under both physiological and pathological conditions remains unclear. RESULTS We utilized an adeno-associated virus gene expression system to induce neuron-specific overexpression of CD47 in wild-type and 5xFAD mice, assessing its effects on microglial synaptic phagocytosis and mouse behaviors. Our results indicate that neuronal CD47 induces ASD-like behaviors and synaptic pruning defects, while promoting behavioral disinhibition and improving memory in wild-type mice. Single-nucleus RNA sequencing was employed to profile gene expression patterns in subpopulations of neurons and microglia. Notably, neuronal CD47 enhances synaptic pathways in neurons and particularly shifts microglial subpopulations from a disease-associated state to a homeostatic state. Additionally, neuronal CD47 reduces excessive microglial synaptic phagocytosis induced by Aβ pathology in 5xFAD mice. CONCLUSION Our study provides evidence that neuronal CD47 overexpression results in synaptic pruning defects and is involved in the pathogenesis of ASD, while also playing a beneficial role in mitigating excessive synaptic loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Mengting Chen
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuxue Lin
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Li Sun
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuping Ye
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yujie Cheng
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Yin Xu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
- Anhui Institute of Translational Medicine, Hefei, 230032, China.
| |
Collapse
|
37
|
Jing X, Zhao Y, Wang G, Tian W. Ginsenoside 1 mitigates postoperative cognitive dysfunction by enhancing microglial Aβ clearance through the endo-lysosomal pathway. Int Immunopharmacol 2025; 150:114281. [PMID: 39946773 DOI: 10.1016/j.intimp.2025.114281] [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: 12/18/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a common complication in patients after surgery, especially in the elderly. The incidence of POCD not only impaired learning and memory, but also increased morbidity and mortality in patients. However, the exploration of therapeutic agents is limited. Ginsenoside 1 (Rg1) is one of the main compounds of ginseng, which exhibits bioactive and neuroprotective efficiency. In the present study, we aimed to investigate the effects of Rg1 on POCD. METHODS The POCD model was established by performing aseptic laparotomy surgery under anesthesia in 18-month mice. The cognition and anxiety of mice were assessed with MWM, OFT, and NOR tests. An in vitro model was performed on BV2 microglial cells. RNA sequencing, Western blotting, electrophysiology, Golgi staining, engulfment, and immunofluorescence analysis were performed. RESULTS Our results showed that Rg1 effectively alleviated the cognitive dysfuncion and anxiety of POCD mice. Transcriptomic sequencing data in microglia indicated that Rg1 mainly affects endosomes and lysosomes. By upregulating Rab7 and TFEB expression, Rg1 promoted microglial engulfment of Aβ through the endo-lysosomal pathway. Additionally, Rg1 reduced inflammatory levels, increased synaptic plasticity, and mitigated neuronal damage caused by Aβ. Moreover, the effects of Rg1 on TFEB depended on MEK/ERK signaling, while activation of MEK reversed Rg1's protective effects. CONCLUSIONS In conclusion, our study demonstrates that Rg1 can effectively ameliorate cognitive and synaptic deficit by enhancing microglial Aβ clearance through the endo-lysosomal pathway in aged POCD mice, which provides a potential strategy for the prevention of POCD.
Collapse
Affiliation(s)
- Xu Jing
- Department of Anesthesiology Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Department of Anesthesiology, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Jiangsu Province 223001, China
| | - Yongchang Zhao
- Department of Anesthesiology, The Affiliated Huaian Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Jiangsu Province 223001, China
| | - Gang Wang
- Department of Anaesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Weiqian Tian
- Department of Anesthesiology Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
38
|
Yi F, Wu H, Zhao HK. Role of triggering receptor expressed on myeloid cells 1/2 in secondary injury after cerebral hemorrhage. World J Clin Cases 2025; 13:100312. [PMID: 40144485 PMCID: PMC11670023 DOI: 10.12998/wjcc.v13.i9.100312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/26/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a common severe emergency in neurosurgery, causing tremendous economic pressure on families and society and devastating effects on patients both physically and psychologically, especially among patients with poor functional outcomes. ICH is often accompanied by decreased consciousness and limb dysfunction. This seriously affects patients' ability to live independently. Although rapid advances in neurosurgery have greatly improved patient survival, there remains insufficient evidence that surgical treatment significantly improves long-term outcomes. With in-depth pathophysiological studies after ICH, increasing evidence has shown that secondary injury after ICH is related to long-term prognosis and that the key to secondary injury is various immune-mediated neuroinflammatory reactions after ICH. In basic and clinical studies of various systemic inflammatory diseases, triggering receptor expressed on myeloid cells 1/2 (TREM-1/2), and the TREM receptor family is closely related to the inflammatory response. Various inflammatory diseases can be upregulated and downregulated through receptor intervention. How the TREM receptor functions after ICH, the types of results from intervention, and whether the outcomes can improve secondary brain injury and the long-term prognosis of patients are unknown. An analysis of relevant research results from basic and clinical trials revealed that the inhibition of TREM-1 and the activation of TREM-2 can alleviate the neuroinflammatory immune response, significantly improve the long-term prognosis of neurological function in patients with cerebral hemorrhage, and thus improve the ability of patients to live independently.
Collapse
Affiliation(s)
- Fan Yi
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
| | - Hao Wu
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an 710038, Shaanxi Province, China
| | - Hai-Kang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Xi’an Medical University, Xi’an 710038, Shaanxi Province, China
| |
Collapse
|
39
|
Walsh AE, Lukens JR. Harnessing microglia-based cell therapies for the treatment of neurodegenerative diseases. Curr Opin Immunol 2025; 94:102552. [PMID: 40138748 DOI: 10.1016/j.coi.2025.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Given the growing evidence linking microglia to the onset and progression of various neurodegenerative diseases, these brain-resident macrophages have emerged as a promising cell type for targeted therapeutic interventions. This review highlights recent studies that utilized innovative, microglia-focused strategies for the treatment of diverse neurodegenerative disorders including lysosomal storage disorders, granulin frontotemporal dementia, and Alzheimer's disease. Cutting-edge therapeutic strategies range from replacing faulty microglia with peripheral macrophage precursors or induced human pluripotent stem cell-derived microglia to engineering microglia that target toxic aggregates or deliver remediating payloads. We also examine the potential limitations as well as the clinical benefits of these strategies.
Collapse
Affiliation(s)
- Adeline E Walsh
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA; Molecular Physiology and Biological Physics Graduate Program, UVA, Charlottesville, VA 22908, USA; Biotechnology Training Program, UVA, 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; Molecular Physiology and Biological Physics Graduate Program, UVA, Charlottesville, VA 22908, USA; Biotechnology Training Program, UVA, Charlottesville, VA 22908, USA.
| |
Collapse
|
40
|
Shi Y, Hu Y, Gan Y, Mi Z, Luo S, Lei J, Fang Q, Li H. Tabersonine ameliorates depressive-like behavior by inhibiting NLRP3 inflammasome activation in a mouse model. Neuropharmacology 2025; 273:110432. [PMID: 40147640 DOI: 10.1016/j.neuropharm.2025.110432] [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: 01/12/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Depression, a common mental disorder, is intimately linked to neuroinflammation. In the central nervous system, microglia, the principal cells involved in immunity, are crucial in neuroinflammation and closely associated with the pathogenesis of depression. Several studies have demonstrated that depressive-like behaviors could be ameliorated by improving brain inflammation. Notably, natural products occupy a critical position in the study of antidepressants. Herein, we explored the antidepressant effects of tabersonine (Tab), a natural inhibitor of NLRP3. Tab significantly improved depressive-like behaviors and anxiety in lipopolysaccharide (LPS)-treated mice. To further elucidate mechanisms underlying the antidepressant actions of Tab, BV2 microglial cells were exposed to LPS and ATP in vitro. Tab effectively inhibited NLRP3 inflammasome activation, subsequent Caspase-1 cleavage, and interleukin-1β secretion both in the hippocampi of mice in vivo and BV2 cells in vitro. Additionally, Tab strongly decreased the concentrations of the proinflammatory cytokines interleukin-1β, tumor necrosis factor, and interleukin-6 in BV2 cell culture supernatants and sera of mice. Further studies indicated that Tab improved LPS-induced neuronal loss, as indicated by a significant rise in the quantity of Nissl-positive cells within the hippocampal regions CA1, CA3, and dentate gyrus. Importantly, Tab counteracted the LPS-induced microglial activation in the hippocampus. Our results indicate that Tab significantly improves LPS-triggered depressive-like behaviors and reverses injuries to hippocampal microglia and neurons, implying its potential as a therapeutic agent for depression.
Collapse
Affiliation(s)
- Yan Shi
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Yue Hu
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Yaoxue Gan
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Zhaoyu Mi
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Shuting Luo
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Jia Lei
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Qian Fang
- School of Medical Technology and Translational Medicine, Hunan Normal University, Changsha, 410006, China
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Skull Base Surgery and Neurooncology in Hunan Province, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
41
|
Wu Z, Wang Y, Chen WW, Sun H, Chen X, Li X, Wang Z, Liang W, Wang SY, Luan X, Li Y, Huang S, Liang Y, Zhang J, Chen ZF, Wang G, Gao Y, Liu Y, Wang J, Liu Z, Shi P, Liu C, Lv L, Hou A, Wu C, Yao C, Hong Z, Dai J, Lu Z, Pan F, Chen X, Kettenmann H, Amit I, Speakman JR, Chen Y, Ginhoux F, Cui R, Huang T, Li H. Peripheral nervous system microglia-like cells regulate neuronal soma size throughout evolution. Cell 2025:S0092-8674(25)00192-8. [PMID: 40199320 DOI: 10.1016/j.cell.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025]
Abstract
Microglia, essential in the central nervous system (CNS), were historically considered absent from the peripheral nervous system (PNS). Here, we show a PNS-resident macrophage population that shares transcriptomic and epigenetic profiles as well as an ontogenetic trajectory with CNS microglia. This population (termed PNS microglia-like cells) enwraps the neuronal soma inside the satellite glial cell envelope, preferentially associates with larger neurons during PNS development, and is required for neuronal functions by regulating soma enlargement and axon growth. A phylogenetic survey of 24 vertebrates revealed an early origin of PNS microglia-like cells, whose presence is correlated with neuronal soma size (and body size) rather than evolutionary distance. Consistent with their requirement for soma enlargement, PNS microglia-like cells are maintained in vertebrates with large peripheral neuronal soma but absent when neurons evolve to have smaller soma. Our study thus reveals a PNS counterpart of CNS microglia that regulates neuronal soma size during both evolution and ontogeny.
Collapse
Affiliation(s)
- Zhisheng Wu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yiheng Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei-Wei Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hua Sun
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Life Sciences, Henan University, Henan, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiaobo Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zeshuai Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weizheng Liang
- Hebei Provincial Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Xuemei Luan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yijiang Li
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Shangjin Huang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuteng Liang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiaqi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhou-Feng Chen
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, and Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Yun Gao
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yanan Liu
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jun Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Peng Shi
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Anli Hou
- Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Chenglin Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Yao
- The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, China
| | - Zexuan Hong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ji Dai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhonghua Lu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yun Chen
- Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rongfeng Cui
- School of Ecology & State Key Laboratory of Biocontrol, Sun Yat-sen University, Shenzhen, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hanjie Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
42
|
Mazzitelli M, Kiritoshi T, Presto P, Hurtado Z, Antenucci N, Ji G, Neugebauer V. BDNF Signaling and Pain Modulation. Cells 2025; 14:476. [PMID: 40214430 PMCID: PMC11987912 DOI: 10.3390/cells14070476] [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: 02/27/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an important neuromodulator of nervous system functions and plays a key role in neuronal growth and survival, neurotransmission, and synaptic plasticity. The effects of BDNF are mainly mediated by the activation of tropomyosin receptor kinase B (TrkB), expressed in both the peripheral and central nervous system. BDNF has been implicated in several neuropsychiatric conditions such as schizophrenia and anxio-depressive disorders, as well as in pain states. This review summarizes the evidence for a critical role of BDNF throughout the pain system and describes contrasting findings of its pro- and anti-nociceptive effects. Different cellular sources of BDNF, its influence on neuroimmune signaling in pain conditions, and its effects in different cell types and regions are described. These and endogenous BDNF levels, downstream signaling mechanisms, route of administration, and approaches to manipulate BDNF functions could explain the bidirectional effects in pain plasticity and pain modulation. Finally, current knowledge gaps concerning BDNF signaling in pain are discussed, including sex- and pathway-specific differences.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Takaki Kiritoshi
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Zachary Hurtado
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (M.M.); (T.K.); (P.P.); (Z.H.); (N.A.); (G.J.)
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
43
|
Cozza M, Boccardi V. Cognitive frailty: A comprehensive clinical paradigm beyond cognitive decline. Ageing Res Rev 2025; 108:102738. [PMID: 40122397 DOI: 10.1016/j.arr.2025.102738] [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: 11/04/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Cognitive frailty is an emerging concept in research and clinical practice that incorporates both physical frailty and mild cognitive impairment (MCI) or subjective cognitive decline (SCD). Unlike traditional approaches that separate physical frailty and dementia, cognitive frailty treats these domains as interrelated and coexisting, with significant implications for clinical outcomes and predicting cognitive decline. Despite growing recognition of this interrelationship, a dualistic view of physical and cognitive processes persists. The paradigm of cognitive frailty holds promise as a biomarker- like amyloid plaques or neurofibrillary tangles- but with the advantage of identifying risk at a prefrail stage, before clinical signs of MCI or dementia emerge. This review examines the pathophysiological and clinical dimensions of cognitive frailty and promotes for its integration into routine assessments in memory clinics.
Collapse
Affiliation(s)
- Mariagiovanna Cozza
- UOC Intermediate Care-Long term Budrio Hospital, Ausl Bologna, Integration Department, Italy
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, Italy.
| |
Collapse
|
44
|
Yang R, Deng MY, Yang LK, Wang GZ, Ma J, Wen Q, Gao N, Qiao HL. Identification of Cytochrome P450 2E1 as a Novel Target in Neuroinflammation and Development of Its Inhibitor Q11 as a Treatment Strategy. Free Radic Biol Med 2025:S0891-5849(25)00182-0. [PMID: 40122152 DOI: 10.1016/j.freeradbiomed.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Neuroinflammation is implicated in nearly all pathological processes of central nervous system (CNS) diseases. However, no medications specifically targeting neuroinflammation are clinically available, and conventional anti-inflammatory drugs exhibit limited efficacy. Here, we identified cytochrome P450 2E1 (CYP2E1) as a novel therapeutic target in neuroinflammation. Elevated CYP2E1 levels were observed in hippocampal tissues of mouse and rat neuroinflammation models, as well as in LPS-stimulated primary microglia. Genetic ablation of Cyp2e1 improved spatial learning and memory in neuroinflammatory rats and reduced pro-inflammatory cytokine levels in Cyp2e1-deficient microglia. Furthermore, Q11 (1-(4-methyl-5-thiazolyl) ethenone), a novel CYP2E1 inhibitor developed and synthesized in our laboratory, effectively ameliorated Alzheimer's disease-related spatial learning and memory functions and depression-related anxiety-like behaviors in mice/rats. Mechanistically, Q11 attenuated microglial activation, neuronal damage, oxidative stress, and neuroinflammation by suppressing the PI3K/Akt, STAT1/3, and NF-κB signaling pathways. These findings establish CYP2E1 as a druggable target for neuroinflammation and propose Q11 as a promising candidate for treating neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Rui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Lu-Kui Yang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Guan-Zhe Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Jun Ma
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Qiang Wen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Na Gao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China
| | - Hai-Ling Qiao
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, zhengzhou 450001, Henan, China.
| |
Collapse
|
45
|
Zou Y, Zhang J, Chen L, Xu Q, Yao S, Chen H. Targeting Neuroinflammation in Central Nervous System Diseases by Oral Delivery of Lipid Nanoparticles. Pharmaceutics 2025; 17:388. [PMID: 40143051 PMCID: PMC11944764 DOI: 10.3390/pharmaceutics17030388] [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: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Neuroinflammation within the central nervous system (CNS) is a primary characteristic of CNS diseases, such as Parkinson's disease, Alzheimer's disease (AD), amyotrophic lateral sclerosis, and mental disorders. The excessive activation of immune cells results in the massive release of pro-inflammatory cytokines, which subsequently induce neuronal death and accelerate the progression of neurodegeneration. Therefore, mitigating excessive neuroinflammation has emerged as a promising strategy for the treatment of CNS diseases. Despite advancements in drug discovery and the development of novel therapeutics, the effective delivery of these agents to the CNS remains a serious challenge due to the restrictive nature of the blood-brain barrier (BBB). This underscores the need to develop a novel drug delivery system. Recent studies have identified oral lipid nanoparticles (LNPs) as a promising approach to efficiently deliver drugs across the BBB and treat neurological diseases. This review aims to comprehensively summarize the recent advancements in the development of LNPs designed for the controlled delivery and therapeutic modulation of CNS diseases through oral administration. Furthermore, this review addresses the mechanisms by which these LNPs overcome biological barriers and evaluate their clinical implications and therapeutic efficacy in the context of oral drug delivery systems. Specifically, it focuses on LNP formulations that facilitate oral administration, exploring their potential to enhance bioavailability, improve targeting precision, and alleviate or manage the symptoms associated with a range of CNS diseases.
Collapse
Affiliation(s)
- Yuan Zou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Longmin Chen
- Department of Rheumatology and Immunology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Qianqian Xu
- Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, NHC Key Laboratory for Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan 430074, China; (J.Z.); (Q.X.)
| | - Sheng Yao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.Z.); (S.Y.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
46
|
Zhang S, Hu H, Wang X, Xiong C, Asmann YW, Ren Y. Single-cell multiomics reveals disrupted glial gene regulatory programs in Alzheimer's disease via interpretable machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643349. [PMID: 40166228 PMCID: PMC11957018 DOI: 10.1101/2025.03.14.643349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Recent development of single-cell technology across multiple omics platforms has provided new ways to obtain holistic views of cells to study disease pathobiology. Alzheimer's disease (AD) is the most common form of dementia worldwide, yet the detailed understanding of its cellular and molecular mechanisms remains limited. In this study, we analyzed paired single-cell transcriptomic (scRNA-seq) and chromatin accessibility (scATAC-seq) data from the Seattle Alzheimer's Disease Brain Cell Atlas (SEA-AD) Consortium to investigate the molecular mechanisms of AD at a cell-subpopulation-specific resolution focusing on glial cells. We benchmarked various multi-omics integration methods using diverse metrics and built an analytic workflow that enabled effective batch correction and cross-modality alignment, creating a unified cell state space. Through integrative analysis of 26 human brain samples, we uncovered AD-associated gene expression and pathway changes in glial subpopulations and highlighted important transcriptomic and epigenomic signatures via functional inference and interpretable machine learning paradigms, discovering the profound involvement of the Solute Carrier proteins (SLC) family genes in multiple glial cell types. We also identified glial cell-specific regulatory programs mediated by key transcription factors such as JUN and FOSL2 in astrocytes, the Zinc Finger (ZNF) family genes in microglia, and the SOX family of transcription factors in oligodendrocytes. Our study provides a comprehensive workflow and a high-resolution view of how glial regulatory programs are disrupted in AD. Our findings offer novel insights into disease-related changes in gene regulation and suggest potential targets for further research and therapy.
Collapse
|
47
|
Francavilla F, Intranuovo F, La Spada G, Lacivita E, Catto M, Graps EA, Altomare CD. Inflammaging and Immunosenescence in the Post-COVID Era: Small Molecules, Big Challenges. ChemMedChem 2025; 20:e202400672. [PMID: 39651728 DOI: 10.1002/cmdc.202400672] [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: 08/28/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/11/2024]
Abstract
Aging naturally involves a decline in biological functions, often triggering a disequilibrium of physiological processes. A common outcome is the altered response exerted by the immune system to counteract infections, known as immunosenescence, which has been recognized as a primary cause, among others, of the so-called long-COVID syndrome. Moreover, the uncontrolled immunoreaction leads to a state of subacute, chronic inflammatory state known as inflammaging, responsible in turn for the chronicization of concomitant pathologies in a self-sustaining process. Anti-inflammatory and immunosuppressant drugs are the current choice for the therapy of inflammaging in post-COVID complications, with contrasting results. The increasing knowledge of the biochemical pathways of inflammaging led to disclose new small molecules-based therapies directed toward different biological targets involved in inflammation, immunological response, and oxidative stress. Herein, paying particular attention to recent clinical data and preclinical literature, we focus on the role of endocannabinoid system in inflammaging, and the promising therapeutic option represented by the CB2R agonists, the role of novel ligands of the formyl peptide receptor 2 and ultimately the potential of newly discovered monoamine oxidase (MAO) inhibitors with neuroprotective activity in the treatment of immunosenescence.
Collapse
Affiliation(s)
- Fabio Francavilla
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Francesca Intranuovo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Gabriella La Spada
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Enza Lacivita
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Elisabetta Anna Graps
- ARESS Puglia - Agenzia Regionale strategica per la Salute ed il Sociale, Lungomare Nazario Sauro 33, 70121, Bari, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| |
Collapse
|
48
|
Wu SR, Nowakowski TJ. Exploring human brain development and disease using assembloids. Neuron 2025:S0896-6273(25)00128-X. [PMID: 40107269 DOI: 10.1016/j.neuron.2025.02.010] [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: 05/08/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
How the human brain develops and what goes awry in neurological disorders represent two long-lasting questions in neuroscience. Owing to the limited access to primary human brain tissue, insights into these questions have been largely gained through animal models. However, there are fundamental differences between developing mouse and human brain, and neural organoids derived from human pluripotent stem cells (hPSCs) have recently emerged as a robust experimental system that mimics self-organizing and multicellular features of early human brain development. Controlled integration of multiple organoids into assembloids has begun to unravel principles of cell-cell interactions. Moreover, patient-derived or genetically engineered hPSCs provide opportunities to investigate phenotypic correlates of neurodevelopmental disorders and to develop therapeutic hypotheses. Here, we outline the advances in technologies that facilitate studies by using assembloids and summarize their applications in brain development and disease modeling. Lastly, we discuss the major roadblocks of the current system and potential solutions.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
49
|
Zheng MY, Luo LZ. The Role of IL-17A in Mediating Inflammatory Responses and Progression of Neurodegenerative Diseases. Int J Mol Sci 2025; 26:2505. [PMID: 40141149 PMCID: PMC11941770 DOI: 10.3390/ijms26062505] [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: 12/16/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
IL-17A has been implicated as a critical pro-inflammatory cytokine in the pathogenesis of autoimmune and neurodegenerative disorders. Emerging evidence indicates its capacity to activate microglial cells and astrocytes, subsequently inducing the production of inflammatory mediators that exacerbate neuronal injury and functional impairment. Clinical observations have revealed a demonstrated association between IL-17A concentrations and blood-brain barrier (BBB) dysfunction, creating a pathological feedback loop that amplifies neuro-inflammatory responses. Recent advances highlight the cytokine's critical involvement in neurodegenerative disorders through multiple molecular pathways. Therapeutic interventions utilizing monoclonal antibodies (mAbs) against IL-17A or its cognate receptor (IL-17R) have shown promising clinical potential. This review systematically examines the IL-17A-mediated neuro-inflammatory cascades; the mechanistic contributions to neurodegenerative pathology in the established disease models including multiple sclerosis, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis; and current therapeutic strategies targeting the IL-17A signaling pathways. The analysis provides novel perspectives on optimizing cytokine-directed therapies while identifying the key challenges and research priorities for translational applications in neurodegeneration.
Collapse
Affiliation(s)
- Miao-Yan Zheng
- School of Pharmacy, Fujian Medical University, Xuefu North Road 1, University Town, Fuzhou 350122, China;
| | - Lian-Zhong Luo
- School of Pharmacy, Fujian Medical University, Xuefu North Road 1, University Town, Fuzhou 350122, China;
- Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical Resources, Xiamen Medical College, 1999 Guankouzhong Road, Xiamen 361023, China
| |
Collapse
|
50
|
Tomer S, Pan L, Harding J, Rezek V, Krawczyk M, Cook E, Kedia N, Wang L, Le J, Martin H, Gramajo E, Li X, Zhang Y, Mu W, Wen J, Kitchen S, Zhang Y, Zhen A. A Novel Glia-immune humanized mouse model for investigation of HIV CNS infection and neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.05.641678. [PMID: 40161608 PMCID: PMC11952312 DOI: 10.1101/2025.03.05.641678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Although combined antiretroviral therapy (ART) is successful in suppressing viral replication, HIV persists in anatomic reservoirs, including the central nervous system (CNS). Current models, such as HSC-reconstituted humanized mice, lack matched human glia in the brain, limiting insights into HIV CNS infection and pathogenesis. We developed a novel glia-immune humanized mouse model integrating human glial cells in the brain with donor-matched human immune reconstitution in peripheral blood and lymphoid tissues. Neonatal NSG mice were injected intrahepatically with Hematopoietic Stem Cells (HSCs) and intracranially with donor-matched glia. We observed extensive engraftment of all three types of human glial cells (astrocytes, oligodendroglia, and microglia-like cells) in key CNS regions, including the cerebral cortex and hippocampus. These glia-immune mice supported robust HIV-1 replication in the peripheral blood, lymphoid tissues and CNS. HIV-infected mice exhibited heightened inflammation and elevated expression of type I interferon-stimulated genes (ISGs) in peripheral and brain tissues. Bulk RNAseq revealed significant transcriptional changes in human glia, such as upregulation of ISGs, inflammasome-associated genes, and downregulation of transcriptional regulators implicated in metabolic regulations and epigenetic controls. Overall, this model allows interrogation of glial transcriptomic changes and neuron-immune interactions, offering insights into HIV CNS infection, pathology, and therapeutic strategies targeting CNS HIV reservoirs and neuroinflammatory pathways.
Collapse
|