1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Tian L, Long G, Zhu S, Wang Y, Xu P, Liu L, Yao H, Fang S, Chen S, Li S. Nanoimmunomodulation of the Aβ-STING feedback machinery in microglia for Alzheimer's disease treatment. Proc Natl Acad Sci U S A 2025; 122:e2427257122. [PMID: 40434641 DOI: 10.1073/pnas.2427257122] [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: 12/31/2024] [Accepted: 04/08/2025] [Indexed: 05/29/2025] Open
Abstract
Imbalanced production and clearance of amyloid-β (Aβ) is a hallmark pathological feature of Alzheimer's disease (AD). While several monoclonal antibodies targeting Aβ have shown reductions in amyloid burden, their impact on cognitive function remains controversial, with the added risk of inflammatory side effects. Dysregulated stimulator of interferon genes (STING) signaling is implicated in neurodegenerative disorders, yet the biological interaction between this pathway and Aβ, as well as their combined influence on AD progression, is poorly understood. Here, we show that while microglia play a protective role in clearing extracellular Aβ, excessive Aβ engulfment triggers the cytosolic leakage of mitochondrial DNA for cGAS-STING cascade. This creates a negative feedback loop that not only exacerbates neuroinflammation but also impairs further Aβ clearance. To address this, we present a nanomedicine approach termed "Aβ-STING Synergistic ImmunoSilencing Therapy (ASSIST)". ASSIST comprises STING inhibitors encapsulated within a blood-brain barrier (BBB)-permeable polymeric micelle that also serves as an Aβ scavenger. Through a multivalent interaction mechanism, ASSIST efficiently destabilizes Aβ plaques and prevents monomer aggregation, subsequently promoting the engulfment of the dissociated Aβ by microglia rather than neurocytes. Furthermore, the STING signaling induced by excessive Aβ uptake is blocked, reducing inflammation and restoring microglial homeostatic functions involved in Aβ clearance. Intravenous administration of ASSIST significantly reduces Aβ burden and improves cognition in AD mice, with minimal cerebral amyloid angiopathy or microhemorrhages. We provide a proof-of-concept nanoengineering strategy to target the maladaptive immune feedback loop arising from conventional immunotherapy for AD treatment.
Collapse
Affiliation(s)
- Limin Tian
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangyu Long
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Siqi Zhu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yuelong Wang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Pengcheng Xu
- Department of Pharmaceutical Engineering, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, China
| | - Lifeng Liu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Yao
- Department of Medicinal Chemistry, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shentong Fang
- School of Biopharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuqing Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Suxin Li
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing 210009, China
| |
Collapse
|
3
|
Li J, He Z, Chai W, Tian M, Yu H, He X, Zhu X. Dip2a regulates stress susceptibility in the basolateral amygdala. Neural Regen Res 2025; 20:1735-1748. [PMID: 39104112 PMCID: PMC11688567 DOI: 10.4103/nrr.nrr-d-23-01871] [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: 11/13/2023] [Revised: 01/31/2024] [Accepted: 03/15/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00025/figure1/v/2024-08-05T133530Z/r/image-tiff Dysregulation of neurotransmitter metabolism in the central nervous system contributes to mood disorders such as depression, anxiety, and post-traumatic stress disorder. Monoamines and amino acids are important types of neurotransmitters. Our previous results have shown that disco-interacting protein 2 homolog A (Dip2a) knockout mice exhibit brain development disorders and abnormal amino acid metabolism in serum. This suggests that DIP2A is involved in the metabolism of amino acid-associated neurotransmitters. Therefore, we performed targeted neurotransmitter metabolomics analysis and found that Dip2a deficiency caused abnormal metabolism of tryptophan and thyroxine in the basolateral amygdala and medial prefrontal cortex. In addition, acute restraint stress induced a decrease in 5-hydroxytryptamine in the basolateral amygdala. Additionally, Dip2a was abundantly expressed in excitatory neurons of the basolateral amygdala, and deletion of Dip2a in these neurons resulted in hopelessness-like behavior in the tail suspension test. Altogether, these findings demonstrate that DIP2A in the basolateral amygdala may be involved in the regulation of stress susceptibility. This provides critical evidence implicating a role of DIP2A in affective disorders.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Zixuan He
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Weitai Chai
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Meng Tian
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Huali Yu
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Xiaoxiao He
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics, Ministry of Education; Institute of Genetics and Cytology, Northeast Normal University, Changchun, Jilin Province, China
| |
Collapse
|
4
|
Zhou Y, Yu G, Li H. Hancinone possesses potentials on increasing the ability of HMC3 cells to phagocytosis of Aβ1-42 via TREM2/Syk/PI3K/AKT/mTOR signaling pathway. PLoS One 2025; 20:e0324202. [PMID: 40424233 DOI: 10.1371/journal.pone.0324202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
CONTEXT The amyloid hypothesis is the most widely accepted explanation for Alzheimer's disease (AD). Failure of microglia Amyloid β-protein (1-42) (Aβ1-42) oligomer clearance and secondary neuroinflammation play a crucial role in the etiology in sporadic AD. Piper kadsura (Choisy) Ohwi (PkO), an herb of Chinese medicine, has anti-inflammation, antioxidation effects. OBJECTIVE To explore the impact of PkO and its active substances on Alzheimer's disease. MATERIALS AND METHODS We integrated drug prediction, network pharmacology and molecular docking techniques to systematically examine multi-scale mechanisms of PkO. Moreover, human Microglia Clone 3 (HMC3) were respectively incubated for 24 hours in the presence or absence of Syk inhibitor (SI, 100 nmol/L), β-amyloid (1-42) oligomer mixtures (called as Aβ oligomer hereafter, Aβ, 2.5 µM), or hancinone (HAN, 0.5 µM, 2.5 µM, 10 µM) to verify the target of the effect of PkO on Aβ oligomer-induced microglia. RESULTS Ultimately, we screened hancinone from PkO as a potential therapeutic agent for AD. Hancinone increased Triggering receptor expressed on myeloid cells 2 (TREM2), Syk, and p-Syk levels, up-regulated relative levels of p-PI3K, p-AKT, and mTOR, promoted the ability of HMC3 cells from the M1 phenotype to the M2 phenotype in Aβ or SI-stimulated HMC3 cells, and enhanced the phagocytic capacity of HMC3 cells to Aβ. DISCUSSION AND CONCLUSIONS Hancinone could regulate the phenotype of HMC3 cells and promote cell phagocytosis of Aβ by modulating the TREM2/Syk/PI3K/AKT/mTOR signaling pathway. This systematic exploration indicates that hancinone has the therapeutic effect on AD.
Collapse
Affiliation(s)
- Yushun Zhou
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Guran Yu
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Hao Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, PR China
| |
Collapse
|
5
|
Choi J, Strickland A, Loo HQ, Dong W, Barbar L, Bloom AJ, Sasaki Y, Jin SC, DiAntonio A, Milbrandt J. Diverse cell types establish a pathogenic immune environment in peripheral neuropathy. J Neuroinflammation 2025; 22:138. [PMID: 40410792 PMCID: PMC12100903 DOI: 10.1186/s12974-025-03459-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Neuroinflammation plays a complex and context-dependent role in many neurodegenerative diseases. We identified a key pathogenic function of macrophages in a mouse model of a rare human congenital neuropathy in which SARM1, the central executioner of axon degeneration, is activated by hypomorphic mutations in the axon survival factor NMNAT2. Macrophage depletion blocked and reversed neuropathic phenotypes in this sarmopathy model, revealing SARM1-dependent neuroimmune mechanisms as key drivers of disease pathogenesis. In this study, we investigated the impact of chronic subacute SARM1 activation on the peripheral nerve milieu using single cell/nucleus RNA-sequencing (sc/snRNA-seq). Our analyses reveal an expansion of immune cells (macrophages and T lymphocytes) and repair Schwann cells, as well as significant transcriptional alterations to a wide range of nerve-resident cell types. Notably, endoneurial fibroblasts show increased expression of chemokines (Ccl9, Cxcl5) and complement components (C3, C4b, C6) in response to chronic SARM1 activation, indicating enhanced immune cell recruitment and immune response regulation by non-immune nerve-resident cells. Analysis of CD45+ immune cells in sciatic nerves revealed an expansion of an Il1b+ macrophage subpopulation with increased expression of markers associated with phagocytosis and T cell activation/proliferation. We also found a significant increase in T cells in sarmopathic nerves. Remarkably, T cell depletion rescued motor phenotypes in the sarmopathy model. These findings delineate the significant changes chronic SARM1 activation induces in peripheral nerves and highlights the potential of immunomodulatory therapies for SARM1-dependent peripheral neurodegenerative disease.
Collapse
Affiliation(s)
- Julie Choi
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hui Qi Loo
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Wendy Dong
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Lilianne Barbar
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - A Joseph Bloom
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA
| | - Yo Sasaki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
6
|
Shen T, Tai W, Jiang D, Ma S, Zhong X, Zou Y, Zhang CL. GADD45G operates as a pathological sensor orchestrating reactive gliosis and neurodegeneration. Neuron 2025:S0896-6273(25)00345-9. [PMID: 40409253 DOI: 10.1016/j.neuron.2025.04.033] [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: 09/05/2024] [Revised: 03/28/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
Reactive gliosis is a hallmark of neuropathology and offers a potential target for addressing numerous neurological diseases. Here, we show that growth arrest and DNA damage inducible gamma (GADD45G), a stress sensor in astrocytes, is a nodal orchestrator of reactive gliosis and neurodegeneration. GADD45G expression in astrocytes is sufficient to incite astrogliosis, microgliosis, synapse loss, compromised animal behavior, and the aggravation of Alzheimer's disease (AD). Conversely, silencing GADD45G specifically in astrocytes preserves synapses and rescues the histological and behavioral phenotypes of AD. Mechanistically, GADD45G controls the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) and neuroimmune signaling pathways, including nuclear factor κB (NF-κB) and interferon regulatory factor 3 (IRF3), leading to profound molecular changes and the secretion of various factors that regulate both cell-autonomous and cell-nonautonomous reactive gliosis and glia-neuron interactions. These results uncover GADD45G signaling as a promising therapeutic target for AD and potentially for numerous other neurological disorders.
Collapse
Affiliation(s)
- Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dongfang Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuaipeng Ma
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
7
|
Nakamizo S, Kabashima K. Cutaneous granulomas: mechanisms, cellular interactions and therapeutic insights. Br J Dermatol 2025; 192:974-982. [PMID: 40080709 DOI: 10.1093/bjd/ljaf096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
Granulomas are specialized biologic defence mechanisms that form in response to infections by pathogens, foreign bodies or specific stimuli such as antimicrobials or fungi. These structures function to isolate foreign materials and pathogens that cannot be eliminated by immune cells, primarily through macrophage activity. In the skin, granulomas are a hallmark of several conditions, including sarcoidosis, granuloma annulare, tuberculosis and leprosy, each exhibiting distinct pathological and immunological features. Granulomas can also arise from lipid accumulation, as observed in xanthogranuloma, or be triggered by inflammatory processes associated with unidentified antigens. Among their cellular components, Langhans-type multinucleated giant cells play a pivotal role in granuloma structure and function, contributing to pathogen containment and tissue remodelling, although their precise mechanisms of action remain an area of active investigation. In addition to these giant cells, recent studies have identified triggering receptors expressed on myeloid cells 2 (TREM2)+ macrophages as key contributors to granuloma formation and maintenance. These macrophages are involved in extracellular degradation of foreign substances and play a role in adapting to the hypoxic and nutrient-poor microenvironment of granulomas through metabolic reprogramming, including the pentose phosphate pathway. Recent advances in molecular biology, such as single-cell RNA sequencing, have provided unprecedented insights into the cellular heterogeneity and molecular pathways involved in granuloma formation. These techniques have revealed disease-specific differences in immune cell profiles and activation states, offering new perspectives on the underlying mechanisms of granulomatous diseases. Despite these advances, the precise processes driving granuloma formation and their functional significance remain largely unclear. This review addresses the central question, 'What is a granuloma?', by synthesizing recent findings, with a particular focus on cutaneous granulomas, and presenting interpretations grounded in the current body of literature. We also discuss the implications of these findings for the development of novel therapeutic strategies, including targeted immunomodulation and cytokine blockade, which hold promise for treating granulomatous diseases while preserving host defence.
Collapse
Affiliation(s)
- Satoshi Nakamizo
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- A*STAR Skin Research Labs (A*SRL), Singapore Immunology Network (SIgN), and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology, and Research (A*STAR), Singapore
| |
Collapse
|
8
|
Zhu W, Zhou Y, Wang Y, Guo L, Liu C. TREM2 in cardiovascular diseases: Mechanisms and therapeutic perspectives. Ageing Res Rev 2025; 109:102774. [PMID: 40381736 DOI: 10.1016/j.arr.2025.102774] [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: 02/21/2025] [Revised: 05/04/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality, with immune responses playing a central role in their pathogenesis. Triggering receptor expressed on myeloid cells 2 (TREM2) is a key immune regulator in CVDs, influencing inflammation, lipid metabolism, and tissue repair. This review comprehensively examines TREM2's structure, function, and signaling pathways, highlighting its roles in atherosclerosis, myocardial infarction, hypertension, atrial fibrillation, and heart failure. In atherosclerosis, macrophages with high TREM2 expression (TREM2hi macrophages) promote plaque progression in early stages but enhance plaque stability in advanced stages. In myocardial infarction, TREM2 modulates macrophage diversity and efferocytosis, aiding cardiac repair. TREM2 also plays a protective role in hypertensive heart disease by reducing inflammation and promoting tissue repair. Challenges in targeting TREM2 therapeutically include its context-dependent effects and complex signaling pathways. Future research should focus on elucidating TREM2's mechanisms in CVDs and developing stage-specific therapies.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| | - Yue Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China.
| | - Yufan Wang
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, PR China.
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, PR China.
| |
Collapse
|
9
|
Higashi Y, Ikuno K, Saito T, Saido TC, Miyasaka T, Kakuda N, Funamoto S. High Affinity Staining for Histological Immunoreactivity revealed phosphorylated tau within amyloid-cored plaques in the brain of AD model mice. Biochem Biophys Res Commun 2025; 771:152025. [PMID: 40393159 DOI: 10.1016/j.bbrc.2025.152025] [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: 05/07/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
The historical pathology of the brain in Alzheimer's disease (AD) is characterized by the amyloid cascade hypothesis, in which amyloid β protein accumulates in the extracellular parenchyma as senile plaque and triggers phosphorylation of microtubule-associated protein tau for forming neurofibrillary tangle in the human neurons. Whether these protein existences differed in the brain parenchyma, the relationship of these proteins of accumulation mechanisms is unknown. In the case of brain pathological analysis, the level of phosphorylation for tau has been decreased in the paraffin-embedded sections compared with biochemical analysis. Here, we have established and developed a method to highlight phosphorylated proteins including tau with frozen sections, as the HIGh Affinity Staining Histological Immunoreactivity (HIGASHI) method. Using this HIGASHI method, hyper-phosphorylated tau could be detected in the mossy fiber on the frozen brain sections of wild-type mice under hypothermia conditions. Here, we attempted the HIGASHI method to detect senile plaque and phosphorylated tau in the AD model mouse brains. Phosphorylated tau was found in the center of senile plaques in the mice brain parenchyma. Additionally, these senile plaques colocalized with microglia cells in the center of senile plaques. Interestingly, senile plaques have been made in the tau knock-out mice brains expressing human amyloid precursor protein. Thus, senile plaques have been composed of Aβ and phosphorylated tau in the brain, but tau isn't necessary for bearing senile plaques.
Collapse
Affiliation(s)
- Yuto Higashi
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Kanta Ikuno
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8602, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Tomohiro Miyasaka
- Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Laboratory of Physiology and Anatomy, Nihon University, School of Pharmacy, Funabashi, Chiba, 274-8555, Japan.
| | - Nobuto Kakuda
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan.
| | - Satoru Funamoto
- Department of Neuropathology, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan; Center for Research in Neurodegenerative Diseases, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan.
| |
Collapse
|
10
|
Jiang M, Li Q, Chen J, Li R, Yao J, Hu Y, Zhang H, Cai L, Luo M, Sun Y, Zeng W. Microglial MS4A4A Protects against Epileptic Seizures in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2417733. [PMID: 40349168 DOI: 10.1002/advs.202417733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/16/2025] [Indexed: 05/14/2025]
Abstract
Alzheimer's disease (AD) is a predominant neurodegenerative disorder worldwide, with epileptic seizures being a common comorbidity that can exacerbate cognitive deterioration in affected individuals, thus highlighting the importance of early therapeutic intervention. It is determined that deletion of Ms4a4a, an AD-associated gene, exacerbates seizures in amyloid β (Aβ)-driven AD mouse model. MS4A4A is significantly upregulated in brain lesions in patients with epilepsy. Single-cell sequencing reveals that MS4A4A is highly expressed in microglia within these lesions, linked to enhanced phagocytic activity. Mechanistic investigation delineates that deletion of Ms4a4a impairs microglial phagocytosis, accompanied by diminished calcium influx and disruptions in mitochondrial metabolic fitness. The cytosolic fragment of Ms4a4a is anchored to the cytoskeletal components, supporting its critical role in mediating phagocytosis. Induction of Ms4a4a through central delivery of LNP-Il4 alleviates seizure conditions. Collectively, these findings identify Ms4a4a as a potential therapeutic target for managing seizures in AD treatment.
Collapse
Affiliation(s)
- Meng Jiang
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Qingqing Li
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Jianhui Chen
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
| | - Ruochong Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Yao
- ENO Bio mRNA Innovation Institute, Shenzhen Rhegen Biotechnology Co. Ltd, Shenzhen, 518000, China
| | - Yong Hu
- ENO Bio mRNA Innovation Institute, Shenzhen Rhegen Biotechnology Co. Ltd, Shenzhen, 518000, China
| | - Haizheng Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lixin Cai
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, 100034, China
| | - Maoguo Luo
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yu Sun
- Pediatric Epilepsy Center, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Zeng
- Institute for Immunology and School of Basic Medical Sciences and Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing, 100084, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Taiyuan, 030001, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| |
Collapse
|
11
|
Xie L, Sheehy RN, Muneer A, Xiong Y, Wrobel JA, Zhang F, Park KS, Velez J, Liu J, Luo YJ, Asrican B, Dong P, Li YD, Damian C, Quintanilla L, Li Y, Xu C, Deshmukh M, Coleman LG, Ming GL, Song H, Wen Z, Jin J, Song J, Chen X. Development of a brain-penetrant G9a methylase inhibitor to target Alzheimer's disease-associated proteopathology. Nat Commun 2025; 16:4222. [PMID: 40328756 PMCID: PMC12056044 DOI: 10.1038/s41467-025-59128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Current Aβ-targeting therapeutics for Alzheimer's disease (AD) only slow cognitive decline due to poor understanding of AD pathogenesis. Here we describe a mechanism of AD pathogenesis in which the histone methyltransferase G9a noncanonically regulates translation of hippocampal proteins associated with AD pathology. Correspondingly, we developed a brain-penetrant inhibitor of G9a, MS1262, which restored both age-related learning & memory and noncognitive functions in multiple AD mouse models. Further, comparison of AD pathology-correlated mouse proteomes with those of AD patients found G9a regulates pathological pathways that promote Aβ and neurofibrillary tangles. This mouse-to-human overlap of G9a regulated AD-associated pathologic proteins supports at the molecular level the efficacy of targeting G9a translational mechanism for treating AD patients. Additionally, MS1262 treatment reversed the AD-characteristic expression or phosphorylation of multiple clinically validated biomarkers of AD that have the potential to be used for early-stage AD diagnosis and companion diagnosis of individualized drug effects.
Collapse
Affiliation(s)
- Ling Xie
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ryan N Sheehy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adil Muneer
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Wrobel
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Velez
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan-Jia Luo
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brent Asrican
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ping Dong
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ya-Dong Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corina Damian
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Luis Quintanilla
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yongyi Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chongchong Xu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Xian Chen
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Cobas-Carreño M, Esteban-Martos A, Tomas-Gallardo L, Iribarren I, Gonzalez-Palma L, Rivera-Ramos A, Elena-Guerra J, Alarcon-Martin E, Ruiz R, Bravo MJ, Venero JL, Morató X, Ruiz A, Royo JL. Drug screening targeting TREM2-TYROBP transmembrane binding. Mol Med 2025; 31:171. [PMID: 40325411 PMCID: PMC12054299 DOI: 10.1186/s10020-025-01229-y] [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: 12/11/2024] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
TREM2 encodes a microglial membrane receptor involved in the disease-associated microglia (DAM) phenotype whose activation requires the transmembrane interaction with TYROBP. Mutations in TREM2 represent a high-impact risk factor for Alzheimer's disease (AD) which turned TREM2 into a significant drug target. We present a bacterial two-hybrid (B2H) system designed for high-throughput screening of modulators for the TREM2-TYROBP transmembrane interaction. In a pilot study, 315 FDA-approved drugs were analyzed to identify potential binding modifiers. Our pipeline includes multiple filtering steps to ensure candidate specificity. The screening suggested two potential candidates that were finally assayed in the human microglial cell line HMC3. Upon stimulation with anti-TREM2 mAb, pSYK/SYK ratios were calculated in the presence of the candidates. As a result, we found that varenicline, a smoking cessation medication, can be considered as a transmembrane agonist of the TREM2-TYROBP interaction.
Collapse
Affiliation(s)
- M Cobas-Carreño
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - A Esteban-Martos
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - L Tomas-Gallardo
- Proteomics and Biochemistry Unit, Andalusian Centre for Developmental Biology, CSIC- Pablo de Olavide, University, Seville, Spain
| | - I Iribarren
- Trinity Biomedical Sciences Institute, School of Chemistry, The University of Dublin, Trinity College, Dublin, Ireland
| | - L Gonzalez-Palma
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - A Rivera-Ramos
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío, CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - J Elena-Guerra
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - E Alarcon-Martin
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - R Ruiz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío, CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - M J Bravo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - J L Venero
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - X Morató
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain
| | - A Ruiz
- Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA.
- Ace Alzheimer Center Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - J L Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, Málaga, Spain.
| |
Collapse
|
13
|
Yagi H, Boeck M, Neilsen K, Yang J, Ko M, Tomita Y, Negishi K, Fu Z, Sun Y, Smith LE. Choroidal Neovascularization Is Suppressed With Activation of TREM2 in Mononuclear Phagocytes-Brief Report. Arterioscler Thromb Vasc Biol 2025; 45:769-777. [PMID: 40143815 PMCID: PMC12017599 DOI: 10.1161/atvbaha.124.321809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [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 real-time quantitative PCR. 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
|
14
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2025; 25:321-352. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
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; 641:718-731. [PMID: 40205047 PMCID: PMC12079183 DOI: 10.1038/s41586-025-08852-z] [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: 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
|
Messenger EJ, Baar SA, Bedford LM, Tsai AP, Lin PB, Ferguson CA, Xu G, Wallace A, Landreth GE, Lamb BT, Bissel SJ. PLCG2 modulates TREM2 expression and signaling in response to Alzheimer's disease pathology. Alzheimers Dement 2025; 21:e70231. [PMID: 40346446 PMCID: PMC12064341 DOI: 10.1002/alz.70231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Phospholipase C gamma 2 (PLCG2) is an intracellular effector of microglial cell surface receptors, including triggering receptor expressed on myeloid cells 2 (TREM2). Variants which alter PLCG2 activity impact Alzheimer's disease (AD) risk, but the effects of PLCG2 deficiency in AD remain unclear. METHODS 5xFAD mice were crossed with PLCG2- and TREM2-deficient mice to assess the role of PLCG2 in response to amyloid pathology. Human bulk RNA-sequencing data were used to validate findings in AD patients. RESULTS In 5xFAD mice, the absence of PLCG2 resulted in reduced TREM2 expression and impaired microglial associations with amyloid beta plaques. Transcriptomic analysis revealed perturbations in immune-related pathways shared between PLCG2 and TREM2 deficiencies, as well as distinct differences. Human transcriptomics revealed positive correlations between PLCG2 and TREM2 independent of pathological scores. DISCUSSION PLCG2 is a critical component of TREM2 signal transduction and may play an upstream role in TREM2 regulation. These findings clarify the mechanisms of risk and protective PLCG2 variants. HIGHLIGHTS The role of phospholipase C gamma 2 (PLCG2) deficiency in response to amyloid beta (Aβ) pathology was investigated in 5xFAD mice and with human cortical transcriptomics. PLCG2 deficiency significantly reduces triggering receptor expressed on myeloid cells 2 (TREM2) expression, while TREM2 deficiency increases PLCG2 expression. PLCG2 expression predicts TREM2 expression in human cortex independent of pathology. PLCG2 and TREM2 deficiencies similarly impair microglial responses to Aβ plaques, exacerbate neuronal pathology, and impair gene expression associated with immune responses. PLCG2 deficiency confers distinct transcriptional perturbations from TREM2 deficiency. PLCG2 may play an upstream role in the regulation of the TREM2-mediated immune response.
Collapse
Affiliation(s)
- Evan J. Messenger
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sydney A. Baar
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Logan M. Bedford
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andy P. Tsai
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Chloe A. Ferguson
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Guixiang Xu
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Abigail Wallace
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gary E. Landreth
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Stephanie J. Bissel
- Stark NeuroscienceIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
17
|
Jocher G, Ozcelik G, Müller SA, Hsia HE, Lastra Osua M, Hofmann LI, Aßfalg M, Dinkel L, Feng X, Schlepckow K, Willem M, Haass C, Tahirovic S, Blobel CP, Lichtenthaler SF. The late-onset Alzheimer's disease risk factor RHBDF2 is a modifier of microglial TREM2 proteolysis. Life Sci Alliance 2025; 8:e202403080. [PMID: 40081988 PMCID: PMC11909414 DOI: 10.26508/lsa.202403080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
The cell surface receptor TREM2 is a key genetic risk factor and drug target in Alzheimer's disease (AD). In the brain, TREM2 is expressed in microglia, where it undergoes proteolytic cleavage, linked to AD risk, but the responsible protease in microglia is still unknown. Another microglial-expressed AD risk factor is catalytically inactive rhomboid 2 (iRhom2, RHBDF2), which binds to and acts as a non-catalytic subunit of the metalloprotease ADAM17. A potential role in TREM2 proteolysis is not yet known. Using microglial-like BV2 cells, bone marrow-derived macrophages, and primary murine microglia, we identify iRhom2 as a modifier of ADAM17-mediated TREM2 shedding. Loss of iRhom2 increased TREM2 in cell lysates and at the cell surface and enhanced TREM2 signaling and microglial phagocytosis of the amyloid β-peptide (Aβ). This study establishes ADAM17 as a physiological TREM2 protease in microglia and suggests iRhom2 as a potential drug target for modulating TREM2 proteolysis in AD.
Collapse
Affiliation(s)
- Georg Jocher
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gozde Ozcelik
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan A Müller
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hung-En Hsia
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Miranda Lastra Osua
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura I Hofmann
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marlene Aßfalg
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lina Dinkel
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Xiao Feng
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kai Schlepckow
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Willem
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Haass
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Sabina Tahirovic
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Carl P Blobel
- Department of Medicine and Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY, USA
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA
- Institute for Advanced Study, Technische Universität München, Garching, Germany
| | - Stefan F Lichtenthaler
- https://ror.org/043j0f473 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- https://ror.org/02kkvpp62 Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
18
|
Yin T, Sun S, Peng L, Yang M, Li M, Yang X, Yuan F, Zhu H, Wang S. Targeting microglial NAAA-regulated PEA signaling counters inflammatory damage and symptom progression of post-stroke anxiety. Cell Commun Signal 2025; 23:211. [PMID: 40312408 PMCID: PMC12046839 DOI: 10.1186/s12964-025-02202-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/12/2025] [Indexed: 05/03/2025] Open
Abstract
Post-stroke anxiety (PSA) manifests as anxiety symptoms after stroke, with unclear mechanisms and limited treatment strategies. Endocannabinoids, reported to mitigate fear, anxiety, and stress, undergo dynamic alterations after stroke linked to prognosis intricately. However, endocannabinoid metabolism in ischemic microenvironment and their associations with post-stroke anxiety-like behavior remain largely uncovered. Our findings indicated that endocannabinoid metabolism was dysregulated after stroke, characterized by elevated N-palmitoylethanolamide (PEA) hydrolase N-acylethanolamine-acid amidase (NAAA) in activated microglia from ischemic area, accompanied by rapid PEA exhaustion. Microglial PEA metabolite exhaustion is directly associated with more severe pathological damage, anxiety symptoms and pain sensitivity. Naaa knockout or pharmacological supplementation to boost PEA pool content can effectively promote stroke recovery and alleviate anxiety-like behaviors. In addition, maintaining PEA pool content in ischemic area reduces overactivated microglia by confronting against mitochondria dysfunction and inflammasome cascade triggered IL-18 release and diffusion to contralateral hemisphere. Meanwhile, maintenance of microglial PEA pool content in ischemic-damaged lesion can preserve contralateral vCA1 synaptic integrity, enhancing anxiolytic pBLA-vCA1Calb1+ circuit activity by alleviating microglial phagocytosis-mediated synaptic loss. Thus, we conclude that microglial NAAA-regulated lipid signaling in the ischemic focus remodels contralateral anxiolytic circuit to participate in post-stroke anxiety progression. Blocking PEA signaling breakdown promotes stroke recovery and mitigates anxiety-like symptoms.
Collapse
Affiliation(s)
- Tianyue Yin
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Shuaijie Sun
- Department of Anesthesiology, Anhui Provincial Hospital, Wannan Medical College, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Mengmeng Yang
- Department of Anesthesiology, Anhui Provincial Hospital, Wannan Medical College, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Mengyu Li
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Xinlu Yang
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Fengyun Yuan
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Core Facility Center, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, 230001, Anhui, China
| | - Hongrui Zhu
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, Division of Life Sciences and Medicine, First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
19
|
Wang NQ, Sun PX, Shen QQ, Deng MY. Cholesterol Metabolism in CNS Diseases: The Potential of SREBP2 and LXR as Therapeutic Targets. Mol Neurobiol 2025; 62:6283-6307. [PMID: 39775479 DOI: 10.1007/s12035-024-04672-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: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
The brain is the organ with the highest cholesterol content in the body. Cholesterol in the brain plays a crucial role in maintaining the integrity of synapses and myelin sheaths to ensure normal brain function. Disruptions in cholesterol metabolism are closely associated with various central nervous system (CNS) diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and multiple sclerosis (MS). In this review, we explore the synthesis, regulation, transport, and functional roles of cholesterol in the CNS. We discuss in detail the associations between cholesterol homeostasis imbalance and CNS diseases including AD, HD, and MS, highlighting the significant role of cholesterol metabolism abnormalities in the development of these diseases. Sterol regulatory element binding protein-2 (SREBP2) and liver X receptor (LXR) are two critical transcription factors that play central roles in cholesterol synthesis and reverse transport, respectively. Their cooperative interaction finely tunes the balance of brain cholesterol metabolism, presenting potential therapeutic value for preventing and treating CNS diseases. We particularly emphasize the alterations in SREBP2 and LXR under pathological conditions and their impacts on disease progression. This review summarizes current therapeutic agents targeting these two pathways, with the hope of broadening the perspectives of CNS drug developers and encouraging further study into SREBP2 and LXR-related therapies for CNS diseases.
Collapse
Affiliation(s)
- Ning-Qi Wang
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Pei-Xiang Sun
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Qi-Qi Shen
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Institute of Clinical Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Meng-Yan Deng
- Institute of Clinical Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
20
|
Wei Z, Pan X, Cui X, Zhang J, Dai X, Zeng Y, Chen X. PU.1 dictates β-amyloid-induced TREM2 expression upregulation in microglia in a transgenic model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1537388. [PMID: 40376093 PMCID: PMC12078285 DOI: 10.3389/fnagi.2025.1537388] [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: 11/30/2024] [Accepted: 04/11/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Microglial dysfunction is characteristic of Alzheimer's disease (AD), with triggering receptor expressed on myeloid cells 2 (TREM2) and transcription factor PU.1 playing crucial roles. However, the relationship between TREM2 and PU.1 remains unclear. Methods We investigated TREM2 and PU.1 expression patterns in the 5×FAD mouse AD model. Experimental approaches included quantitative PCR, western blotting, immunofluorescence staining, chromatin immunoprecipitation, and luciferase reporter assays to examine the interaction between PU.1 and TREM2. The phagocytic function of microglial cells was evaluated using Aβ42 and Nile red fluorescent microsphere phagocytosis assays. Results TREM2 and PU.1 expression significantly correlated with brain β-amyloid (β) deposition. PU.1 directly interacted with the TREM2 promoter region, promoting its transcription and potently impacting microglial phagocytosis. PU.1 overexpression amplified TREM2 expression, while PU.1 knockdown reduced it. Discussion Our findings reveal a novel regulatory mechanism where PU.1 directly modulates TREM2 transcription in activated microglia during AD progression. These insights highlight the potential of TREM2 and PU.1 as therapeutic targets in AD treatment.
Collapse
Affiliation(s)
- Zhen Wei
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
- Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Xiaoli Cui
- Department of Geriatric, The People’s Hospital Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Xiaoman Dai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| | - Yuqi Zeng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou, China
| |
Collapse
|
21
|
Zhang L, Xiang X, Li Y, Bu G, Chen XF. TREM2 and sTREM2 in Alzheimer's disease: from mechanisms to therapies. Mol Neurodegener 2025; 20:43. [PMID: 40247363 PMCID: PMC12004684 DOI: 10.1186/s13024-025-00834-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor predominantly expressed by microglia in the brain. Recent studies have established TREM2 as a central immune signaling hub in neurodegeneration, where it triggers immune responses upon sensing pathological development and tissue damages. TREM2 binds diverse ligands and activates downstream pathways that regulate microglial phagocytosis, inflammatory responses, and metabolic reprogramming. Interestingly, TREM2 exists both in its membrane-bound form and as a soluble variant (sTREM2), that latter is generated through proteolytic shedding or alternative splicing and can be detected in cerebrospinal fluid and plasma. Emerging clinical and preclinical evidence underscores the potential of TREM2 and sTREM2 as diagnostic biomarkers and therapeutic targets in Alzheimer's disease (AD). This review provides a comprehensive overview of the molecular functions, regulatory mechanisms, and pathological implications of TREM2 and sTREM2 in AD. Furthermore, we explore their potential roles in diagnostics and therapeutics while suggesting key research directions for advancing TREM2/sTREM2-based strategies in combating AD.
Collapse
Affiliation(s)
- Lianshuai Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Xianyuan Xiang
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518055, China.
| | - Yahui Li
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Guojun Bu
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xiao-Fen Chen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
22
|
Cai Z, Wang S, Cao S, Chen Y, Penati S, Peng V, Yuede CM, Beatty WL, Lin K, Zhu Y, Zhou Y, Colonna M. Loss of ATG7 in microglia impairs UPR, triggers ferroptosis, and weakens amyloid pathology control. J Exp Med 2025; 222:e20230173. [PMID: 39945772 PMCID: PMC11823820 DOI: 10.1084/jem.20230173] [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: 01/27/2023] [Revised: 11/19/2024] [Accepted: 01/08/2025] [Indexed: 02/16/2025] Open
Abstract
Microglia impact brain development, homeostasis, and pathology. One important microglial function in Alzheimer's disease (AD) is to contain proteotoxic amyloid-β (Aβ) plaques. Recent studies reported the involvement of autophagy-related (ATG) proteins in this process. Here, we found that microglia-specific deletion of Atg7 in an AD mouse model impaired microglia coverage of Aβ plaques, increasing plaque diffusion and neurotoxicity. Single-cell RNA sequencing, biochemical, and immunofluorescence analyses revealed that Atg7 deficiency reduces unfolded protein response (UPR) while increasing oxidative stress. Cellular assays demonstrated that these changes lead to lipoperoxidation and ferroptosis of microglia. In aged mice without Aβ buildup, UPR reduction and increased oxidative damage induced by Atg7 deletion did not impact microglia numbers. We conclude that reduced UPR and increased oxidative stress in Atg7-deficient microglia lead to ferroptosis when exposed to proteotoxic stress from Aβ plaques. However, these microglia can still manage misfolded protein accumulation and oxidative stress as they age.
Collapse
Affiliation(s)
- Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shoutang Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Siyan Cao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Silvia Penati
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Carla M. Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kent Lin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yiyang Zhu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
Fumagalli L, Nazlie Mohebiany A, Premereur J, Polanco Miquel P, Bijnens B, Van de Walle P, Fattorelli N, Mancuso R. Microglia heterogeneity, modeling and cell-state annotation in development and neurodegeneration. Nat Neurosci 2025:10.1038/s41593-025-01931-4. [PMID: 40195564 DOI: 10.1038/s41593-025-01931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
Within the CNS, microglia execute various functions associated with brain development, maintenance of homeostasis and elimination of pathogens and protein aggregates. This wide range of activities is closely associated with a plethora of cellular states, which may reciprocally influence or be influenced by their functional dynamics. Advancements in single-cell RNA sequencing have enabled a nuanced exploration of the intricate diversity of microglia, both in health and disease. Here, we review our current understanding of microglial transcriptional heterogeneity. We provide an overview of mouse and human microglial diversity encompassing aspects of development, neurodegeneration, sex and CNS regions. We offer an insight into state-of-the-art technologies and model systems that are poised to improve our understanding of microglial cell states and functions. We also provide suggestions and a tool to annotate microglial cell states on the basis of gene expression.
Collapse
Affiliation(s)
- Laura Fumagalli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Alma Nazlie Mohebiany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jessie Premereur
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Paula Polanco Miquel
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Baukje Bijnens
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Nicola Fattorelli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
24
|
Islam R, Choudhary HH, Zhang F, Mehta H, Yoshida J, Thomas AJ, Hanafy K. Microglial TLR4-Lyn kinase is a critical regulator of neuroinflammation, Aβ phagocytosis, neuronal damage, and cell survival in Alzheimer's disease. Sci Rep 2025; 15:11368. [PMID: 40175501 PMCID: PMC11965285 DOI: 10.1038/s41598-025-96456-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
Disease-Associated Microglia (DAM) are a focus in Alzheimer's disease (AD) research due to their central involvement in the response to amyloid-beta plaques. Microglial Toll-like receptor 4 (TLR4) is instrumental in the binding of fibrillary amyloid proteins, while Lyn kinase (Lyn) is a member of the Src family of non-receptor tyrosine kinases involved in immune signaling. Lyn is a novel, non-canonical, intracellular adaptor with diverse roles in cell-specific signaling which directly binds to TLR4 to modify its function. Lyn can be activated in response to TLR4 stimulation, leading to phosphorylation of various substrates and modulation of inflammatory and phagocytosis signaling pathways. Here, we investigated the TLR4-Lyn interaction in neuroinflammation using WT, 5XFAD, and 5XFAD x Lyn-/- mouse models by western blotting (WB), co-immunoprecipitation (co-IP), immunohistochemistry (IHC) and flow cytometric (FC) analysis. A spatial transcriptomic analysis of microglia in WT, 5XFAD, and 5XFAD x Lyn-/- mice revealed essential genes involved in neuroinflammation, Aβ phagocytosis, and neuronal damage. Finally, we explored the effects of a synthetic, TLR4-Lyn modulator protein (TLIM) through an in vitro AD model using primary murine microglia. Our WB, co-IP, IHC, and FC data show an increased, novel, direct protein-protein interaction between TLR4 and Lyn kinase in the brains of 5XFAD mice compared to WT. Furthermore, in the absence of Lyn (5XFAD x Lyn-/- mice); increased expression of protective Syk kinase was observed, enhanced microglial Aβ phagocytosis, increased astrocyte activity, decreased neuronal dystrophy, and a further increase in the cell survival signaling and protective DAM population was noted. The DAM population in 5XFAD mice which produce more inflammatory cytokines and phagocytose more Aβ were observed to express greater levels of TLR4 and Lyn. Pathway analysis comparison between WT, 5XFAD, and 5XFAD x Lyn-/- mice supported these findings via our microglial spatial transcriptomic analysis. Finally, we created an in vitro co-culture system with primary murine microglial and primary murine hippocampal cells exposed to Aβ as a model of AD. When these co-cultures were treated with our TLR4-Lyn Interaction Modulators (TLIMs), an increase in Aβ phagocytosis and a decrease in neuronal dystrophy was seen. Lyn kinase has a central role in modulating TLR4-induced inflammation and Syk-induced protection in a 5XFAD mouse model. Our TLIMs ameliorate AD sequalae in an in vitro model of AD and could be a promising therapeutic strategy to treat AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Feng Zhang
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA
| | - Jun Yoshida
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Ajith J Thomas
- Cooper Medical School of Rowan University, Camden, NJ, USA
- Department of Neurosurgery, Cooper University Health Care, Camden, NJ, USA
| | - Khalid Hanafy
- Cooper Medical School of Rowan University, Camden, NJ, USA.
- Department of Neurology, Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School of Rowan University, 401 Broadway, Camden, NJ, 08103, USA.
| |
Collapse
|
25
|
Feng Z, Fu Y, Yang S, Zhao H, Lin M, Liu C, Huang W, He X, Chen Y, Chen J, Shen Y, Li Z, Chen Q. Siglec-15 is a putative receptor for porcine epidemic diarrhea virus infection. Cell Mol Life Sci 2025; 82:136. [PMID: 40172660 PMCID: PMC11965083 DOI: 10.1007/s00018-025-05672-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Porcine epidemic diarrhea virus (PEDV) has caused significant losses in the pork industry, but the mechanism of PEDV infection is still unclear. On the basis of our RNA-Seq data and due to the potential role of sialic acid as a coreceptor, we investigated the function of sialic acid-binding Ig-like lectin 15 (Siglec-15) to determine its role as a receptor in PEDV infection. We found that Siglec-15 enhances PEDV infection by promoting viral adsorption to host cells. Coimmunoprecipitation and immunofluorescence assays revealed that Siglec-15 binds to the S1 subunit and M protein of PEDV. PEDV infectivity was significantly reduced in Siglec-15 knockout mice. In addition, we developed a monoclonal antibody targeting Siglec-15 that can effectively inhibit PEDV infection both in vitro and in vivo. Overall, our study suggests that Siglec-15 may be a receptor for PEDV infection, which is important for related mechanistic studies and reveals a novel target for anti-PEDV therapeutic development.
Collapse
Affiliation(s)
- Zhihua Feng
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, Fujian, 350117, China
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Yajuan Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Sheng Yang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Heng Zhao
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Minhua Lin
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Chuancheng Liu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Weili Huang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Xinyan He
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Yao Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, Fujian, 350117, China
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350117, China
| | - Yangkun Shen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
| | - Zhaolong Li
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian Province, 350013, China.
| | - Qi Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, Fujian, 350117, China.
- College of Photonic and Electronic Engineering, Fujian Normal University, Fuzhou, Fujian, 350117, China.
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou, Fujian Province, 350117, China.
| |
Collapse
|
26
|
Van Hove H, De Feo D, Greter M, Becher B. Central Nervous System Macrophages in Health and Disease. Annu Rev Immunol 2025; 43:589-613. [PMID: 40036702 DOI: 10.1146/annurev-immunol-082423-041334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The central nervous system (CNS) has a unique set of macrophages that seed the tissue early during embryonic development. Microglia reside in the parenchyma, and border-associated macrophages are present in border regions, including the meninges, perivascular spaces, and choroid plexus. CNS-resident macrophages support brain homeostasis during development and steady state. In the diseased brain, however, the immune landscape is altered, with phenotypic and transcriptional changes in resident macrophages and the invasion of blood-borne monocytes, which differentiate into monocyte-derived macrophages upon entering the CNS. In this review, we focus on the fate and function of the macrophage compartment in health, neurodegenerative conditions such as amyloidosis, and neuroinflammation as observed in multiple sclerosis and infection. We discuss our current understanding that monocyte-derived macrophages contribute to neuropathology whereas native macrophages play a neuroprotective role in disease.
Collapse
Affiliation(s)
- Hannah Van Hove
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| |
Collapse
|
27
|
Chen Y, Kou Y, Ni Y, Yang H, Xu C, Fan H, Liu H. Microglia efferocytosis: an emerging mechanism for the resolution of neuroinflammation in Alzheimer's disease. J Neuroinflammation 2025; 22:96. [PMID: 40159486 PMCID: PMC11955113 DOI: 10.1186/s12974-025-03428-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by significant neuroinflammatory responses. Microglia, the immune cells of the central nervous system, play a crucial role in the pathophysiology of AD. Recent studies have indicated that microglial efferocytosis is an important mechanism for clearing apoptotic cells and cellular debris, facilitating the resolution of neuroinflammation. This review summarizes the biological characteristics of microglia and the mechanisms underlying microglial efferocytosis, including the factors and signaling pathways that regulate efferocytosis, the interactions between microglia and other cells that influence this process, and the role of neuroinflammation in AD. Furthermore, we explore the role of microglial efferocytosis in AD from three perspectives: its impact on the clearance of amyloid plaques, its regulation of neuroinflammation, and its effects on neuroprotection. Finally, we summarize the current research status on enhancing microglial efferocytosis to alleviate neuroinflammation and improve AD, as well as the future challenges of this approach as a therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yongping Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Yuhong Kou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Yang Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Cailin Xu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China.
| | - Huanqi Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
28
|
Zhou C, Liang C, Zhang R, Wang Y, Luo S, Pan J. TREM2 improves coagulopathy and lung inflammation in sepsis through the AKT-mTOR pathway. Int Immunopharmacol 2025; 150:114330. [PMID: 39970715 DOI: 10.1016/j.intimp.2025.114330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Sepsis is a systemic inflammatory response syndrome triggered by infection, often accompanied by severe coagulopathy, leading to high mortality. Tissue factor (TF) plays a pivotal role in sepsis by promoting both coagulation and inflammation. Recently, TREM2 (Triggering Receptor Expressed on Myeloid cells 2) has emerged as a key regulator of macrophage function, but its specific role in sepsis remains unclear. METHODS An in vitro sepsis model was established by stimulating RAW264.7 cells with 10 μg/mL lipopolysaccharide (LPS) for 6 h, with four groups: Negative Control (NC), NC + LPS, TREM2, and TREM2 + LPS. Inflammatory cytokines and coagulation factors were measured in each group. Cells in the TREM2 and TREM2 + LPS groups were pretreated with TREM2 overexpression plasmid for 48 h. In vivo, mice were assigned to Sham, TREM2, Cecal Ligation and Puncture (CLP), CLP + NC, and CLP + TREM2 groups. Mice in the NC group received macrophages via tail vein injection, while those in the TREM2 and CLP + TREM2 groups received TREM2-overexpressing macrophages. Lung tissue and plasma samples were collected to assess inflammatory cytokines, coagulation factors, and signaling pathway activity. RESULTS TREM2 overexpression significantly improved survival, reduced lung inflammation, and alleviated coagulopathy in mice. It increased platelet counts and reduced fibrin deposition. Furthermore, TREM2 inhibited TF release from macrophages by suppressing aberrant activation of the AKT-mTOR signaling pathway, thereby modulating the macrophage inflammatory response. CONCLUSIONS TREM2 plays a crucial protective role in sepsis-associated coagulopathy, suggesting that it could serve as a potential therapeutic target, providing novel strategies to improve clinical outcomes in sepsis patients.
Collapse
Affiliation(s)
- Chen Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China
| | - Chenglong Liang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China
| | - Rongrong Zhang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China
| | - Ying Wang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China
| | - Shuang Luo
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingye Pan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Wenzhou Medical University, Wenzhou 325000, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315300, China; Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, 325000, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, 325000, China.
| |
Collapse
|
29
|
Zhai L, Shen H, Wu S, Guo L, Yang Y, Sheng J, Han C. Deer antler polypeptides inhibit microglial activation via TREM2 to improve behavior and neuroinflammation in CUMS mice. Int Immunopharmacol 2025; 150:114284. [PMID: 39955919 DOI: 10.1016/j.intimp.2025.114284] [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: 12/01/2024] [Revised: 01/11/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
To investigate the effects and mechanisms of deer antler total polypeptides (VAP-T) and its active component Y (VAP-Y) in treating depression in CUMS mice.VAP-T improved depression-like behavior in CUMS mice, reduced microglial activation, and tissue inflammation. VAP-Y showed better improvement in depression-like behavior in CUMS mice compared to VAP-T, significantly inhibiting microglial activation and tissue inflammation. Chromatographic analysis of VAP-Y revealed that short peptides had good binding activity with TREM2. VAP-T and VAP-Y have significant improvement effects on depression-like behavior in CUMS mice, which is related to the inhibition of TREM2-mediated microglial activation. VAP-Y has better activity than the total polypeptides and is a polypeptide with potential for treating depression.
Collapse
Affiliation(s)
- Liping Zhai
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Heping Shen
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Shasha Wu
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Li Guo
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Yi Yang
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Jian Sheng
- The Second Affiliated Hospital of Jiaxing University, China.
| | - Chenyang Han
- The Second Affiliated Hospital of Jiaxing University, China.
| |
Collapse
|
30
|
Cao F, Zhang P, Chi Y, Wang Y, Xu L, Zhang D. Celastrol Ameliorated Alzheimer's Disease in Mice by Enhancing TBX21/TREM2 Expression in Microglia and Inhibiting Tau Phosphorylation. Neurochem Res 2025; 50:126. [PMID: 40138096 DOI: 10.1007/s11064-025-04375-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder that is typified by the formation of senile plaques containing Aβ and neurofibrillary tangles containing tau in a hyperphosphorylated state. Celastrol, a natural compound, has proven effective in alleviating AD pathology by enhancing autophagy and reducing tau aggregates. The present study investigates the neuroprotective mechanisms of celastrol, with a particular focus on the participation of the transcription factor T-box transcription factor 21 (TBX21) and triggering receptor expressed on myeloid cells 2 (TREM2) in microglial cells. In AD mouse models, celastrol upregulated TBX21 and TREM2, suppressed phosphorylated tau and inflammatory cytokines, and restored neuronal viability. In vitro, celastrol-treated microglia enhanced neuronal survival under amyloid-beta (Aβ) stress, effects abolished by TBX21/TREM2 knockdown. Mechanistically, TBX21 directly bound the TREM2 promoter to regulate its expression. These findings identified the TBX21-TREM2 axis as a therapeutic target for AD.
Collapse
Affiliation(s)
- Fanfan Cao
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, No. 516, Jungong Rd., Yangpu District, Shanghai, 200093, PR China
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China
| | - Pan Zhang
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China
| | - Yongbin Chi
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China
| | - Ying Wang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China
| | - Limin Xu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China.
| | - Denghai Zhang
- Shanghai Health Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Department of Central Laboratory, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai, 200135, PR China.
| |
Collapse
|
31
|
Dongre P, Ramesh M, Govindaraju T, Inamdar MS. Asrij/OCIAD1 depletion reduces inflammatory microglial activation and ameliorates Aβ pathology in an Alzheimer's disease mouse model. J Neuroinflammation 2025; 22:89. [PMID: 40114191 PMCID: PMC11924864 DOI: 10.1186/s12974-025-03415-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-beta (Aβ) plaques and neurofibrillary tangles, neuroinflammation, and glial activation. Asrij/OCIAD1 (Ovarian Carcinoma Immunoreactive Antigen Domain containing protein 1) is an AD-associated factor. Increased Asrij levels in the brains of AD patients and mouse models are linked to the severity of neurodegeneration. However, the contribution of Asrij to AD progression and whether reducing Asrij levels is sufficient to mitigate Aβ pathology in vivo is unclear. METHODS To explore the impact of Asrij on AD pathology, we deleted asrij in the APP/PS1 mouse model of AD and analyzed the effects on AD hallmarks. We used the Morris water maze and open field test to assess behavioral performance. Using immunohistochemistry and biochemical analyses, we evaluated Aβ plaque load, neuronal and synaptic damage, and gliosis. Further, we utilized confocal microscopy imaging, flow cytometry, and RNA sequencing analysis to comprehensively investigate changes in microglial responses to Aβ pathology upon Asrij depletion. RESULTS Asrij depletion ameliorates cognitive impairments, Aβ deposition, neuronal and synaptic damage, and reactive astrogliosis in the AD mouse. Notably, Asrij-deficient microglia exhibit reduced plaque-associated proliferation and decreased phagocytic activation. Transcriptomic analyses of AD microglia reveal upregulation of energy metabolism pathways and downregulation of innate immunity and inflammatory pathways upon Asrij depletion. Mechanistically, loss of Asrij increases mitochondrial activity and impedes the acquisition of a pro-inflammatory disease-associated microglia (DAM) state. Reduced levels of proinflammatory cytokines and decreased STAT3 and NF-κB activation indicate protective changes in AD microglia. Taken together, our results suggest that increased Asrij levels reported in AD, may suppress microglial metabolic activity and promote inflammatory microglial activation, thereby exacerbating AD pathology. CONCLUSIONS In summary, we show that Asrij depletion ameliorates Aβ pathology, neuronal and synaptic damage, gliosis, and improves behavioral performance in APP/PS1 mice. This supports that Asrij exacerbates the AD pathology. Mechanistically, Asrij is critical for the development of DAM and promotes neuroinflammatory signaling activation in microglia, thus restricting neuroprotective microglial responses. Hence, reducing Asrij in this context may help retard AD. Our work positions Asrij as a critical molecular regulator that links microglial dysfunction to AD pathogenesis.
Collapse
Affiliation(s)
- Prathamesh Dongre
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, 560064, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, 560064, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, 560064, India.
| | - Maneesha S Inamdar
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bangalore, Karnataka, 560064, India.
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, 560065, India.
| |
Collapse
|
32
|
Ma L, Wang J, Zhou R, Chen M, Huang Z, Lin S. Traditional Chinese Medicine-derived formulations and extracts modulating the PI3K/AKT pathway in Alzheimer's disease. Front Pharmacol 2025; 16:1528919. [PMID: 40166467 PMCID: PMC11955602 DOI: 10.3389/fphar.2025.1528919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder characterized by memory decline, cognitive impairment, and behavioral abnormalities. Pathologically, AD is marked by neurofibrillary tangles caused by excessive phosphorylation of Tau protein and abnormal deposition of β-amyloid (Aβ) in the brain. The PI3K/AKT signaling pathway plays a crucial role in the development, survival, and metabolic regulation of the central nervous system, particularly in neuronal growth, differentiation, and apoptosis. However, this pathway is often inhibited in AD patients.In recent years, studies have shown that herbal formulations and extracts derived from Traditional Chinese Medicine (TCM) can regulate the PI3K/AKT signaling pathway, thereby improving AD pathological models. This study reviews fundamental research on both active metabolites and compound formulations from TCM for the treatment of AD, targeting the PI3K/AKT signaling pathway.Keywords include "Alzheimer's disease" "AD" "dementia" "PI3K" "AKT" "Traditional Chinese Medicine" "Chinese herbology" "Chinese medicine" and "TCM".The study is based on relevant literature published over the past 15 years, primarily sourced from electronic databases such as Web of Science, PubMed, CNKI, Wanfang, and VIP databases.The findings indicate that herbal formulations and extracts derived from TCM can mitigate AD pathology by regulating the PI3K/AKT signaling pathway, reducing Tau protein phosphorylation and Aβ deposition, inhibiting inflammatory responses and oxidative stress, and alleviating neuronal apoptosis. This study enhances our understanding of the anti-AD mechanisms of TCM through the PI3K/AKT pathway and offers new insights for the future.
Collapse
Affiliation(s)
- Lan Ma
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Jing Wang
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Rong Zhou
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Miao Chen
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Zuxiu Huang
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Shuyang Lin
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
33
|
Dempsey JL, Savard C, Kothari V, Tang J, Lee SP, Bornfeldt KE, Carr RM, Ioannou GN. Soluble TREM2 is a biomarker but not a mediator of fibrosing steatohepatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642665. [PMID: 40161684 PMCID: PMC11952433 DOI: 10.1101/2025.03.11.642665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Triggering receptor expressed on myeloid cells 2 (TREM2), a transmembrane, lipid-sensing protein expressed by Kupffer cells, is thought to play a role in metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH). Plasma levels of the TREM2 cleavage product, soluble TREM2 (sTREM2), are strongly associated with MASLD severity. We investigated the role of TREM2 in MASH pathogenesis and whether sTREM2 acts both as biomarker and mediator of MASH. Methods Adult C57BL/6J mice were assigned to normal, high-fat (HF), or high-fat and high-cholesterol (HFHC) diets for 15, 30, 90, and 180 days. Plasma sTrem2 levels, liver pathology, and hepatic RNA expression were assessed. To test whether sTrem2 is a mediator of MASH, C57BL/6J mice were injected retro-orbitally with a liver-targeted adeno-associated virus, TBG-AAV8-sTrem2, resulting in secretion of sTrem2 by hepatocytes, or empty TBG-AAV8-control, and subsequently fed HFHC diet for 15, 90, or 180 days. Results HFHC-fed mice developed fibrosing steatohepatitis at 180 days together with a 15-fold increase in plasma sTrem2 levels. In the livers of HFHC-fed mice, crown-like structures (CLSs) consisting of TREM2+ macrophages surrounded necrotic, steatotic hepatocytes and their remnant lipid droplets, which contained prominent crystals containing cholesterol. TBG-AAV8-sTrem2-injected mice had higher levels of plasma sTrem2 than TBG-AAV8-control-injected mice, but there were no differences in liver weight, body weight, hepatic fibrosis, hepatic inflammation, hepatic cholesterol crystals, or plasma cholesterol levels. Conclusion TREM2+ macrophages characterize the CLSs that surround necrotic hepatocytes and their remnant lipid droplets and cholesterol crystals in fibrosing steatohepatitis-MASH. Plasma sTrem2 is a biomarker, but not a causal mediator, of MASH.
Collapse
Affiliation(s)
- Joseph L. Dempsey
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Christopher Savard
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA
| | - Vishal Kothari
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jingjing Tang
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Sum P. Lee
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Karin E. Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Rotonya M. Carr
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - George N. Ioannou
- Division of Gastroenterology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA
- Research and Development, Veterans Affairs Puget Sound Health Care System, Seattle, WA
| |
Collapse
|
34
|
Casali BT, Lin L, Benedict O, Zuppe H, Marsico E, Reed EG. Sex chromosomes and gonads modify microglial-mediated pathology in a mouse model of Alzheimer's disease. J Neuroinflammation 2025; 22:81. [PMID: 40083008 PMCID: PMC11907917 DOI: 10.1186/s12974-025-03404-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder disproportionally affecting women with sex-specific disease manifestations and therapeutic responses. Microglial-mediated inflammation occurs in response to and perpetuates disease processes, and fundamental sex differences in microglia may contribute to these sex biases. Both sex chromosomes and gonad-derived hormones shape immune responses, but their contribution to immune-mediated mechanisms underlying the sex bias in AD is unclear. Crossing the Four Core Genotype (FCG) model to separate sex chromosome and gonad-derived hormone effects to the 5xFAD model, we found the sex chromosome complement impacted microgliosis, neuroinflammation, plaque burden and neuritic dystrophy. Modification of pathology largely correlated with microgliosis, and sex chromosomes and gonad-derived hormones influenced plaque remodeling and microglial CD11c expression. Our results provide potential trajectories for studying and targeting microglial-mediated sex differences and emphasize the complex interplay between sex chromosomes and hormones during AD.
Collapse
Affiliation(s)
- Brad T Casali
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA
| | - Li Lin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA
| | - Olesia Benedict
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA
| | - Hannah Zuppe
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA
| | - Emily Marsico
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA
| | - Erin G Reed
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, 4209 St. Rt. 44, Rootstown, OH, 44272, USA.
| |
Collapse
|
35
|
Zhang X, Wang R, Pan X, Chen Z, Li Y, Hu Y, Liu F, Cao X. TREM2 Modulates Postoperative Cognitive Function in Aged Mice by Inhibiting the NLRP3/caspase-1 Pathway and Apoptosis via PLCγ2 Activation. Mol Neurobiol 2025:10.1007/s12035-025-04820-w. [PMID: 40075039 DOI: 10.1007/s12035-025-04820-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent complication in elderly patients, with neuroinflammation identified as a key contributing factor. This study investigates the therapeutic potential of the TREM2-PLCγ2 signaling pathway in mitigating neuroinflammation, neuronal apoptosis and cognitive impairment following surgery. We employed both in vivo and in vitro models to investigate the effects of TREM2 activation and its interaction with PLCγ2. Mice subjected to surgery were pre-treated with the TREM2-activating peptide COG1410, and subsequently evaluated for neuroinflammation, neuronal apoptosis, and cognitive function. In vitro studies using microglial cells were conducted to examine the mechanistic relationship between TREM2 and PLCγ2 phosphorylation via SYK. Knockdown experiments and SYK inhibition were performed to determine the hierarchical interaction between TREM2, PLCγ2, and their downstream influence on the NLRP3 inflammasome. Surgery significantly elevated the activation of the NLRP3 inflammasome, along with increased Cleaved Caspase-1, IL-1β, IL-18, and neuronal apoptosis markers. Pre-treatment with COG1410 effectively reduced these pro-inflammatory and pro-apoptotic markers, while alleviating cognitive impairment. TREM2 activation promoted SYK-dependent phosphorylation of PLCγ2, which inhibited NLRP3 inflammasome activation and reduced neuroinflammation. TREM2 knockdown exacerbated microglial inflammation, while PLCγ2 knockdown suppressed NLRP3 activation. Inhibition of SYK impaired the protective effects of the TREM2-PLCγ2 pathway and delayed cognitive recovery in mice. RNA-sequencing further revealed significant alterations in pathways related to neuroinflammation and apoptosis. TREM2 modulates PLCγ2 activity through SYK phosphorylation, thereby alleviating microglial-driven neuroinflammation and neuronal apoptosis, and improving cognitive impairment following surgery. Targeting the TREM2-PLCγ2 pathway presents a promising strategy for the prevention and treatment of POCD.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Renyi Wang
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Xue Pan
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Zitong Chen
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Yilong Li
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Yue Hu
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Fang Liu
- Department of Neurology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| | - Xuezhao Cao
- Department of Anesthesiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
36
|
Ye J, Shan F, Xu X, Liang C, Zhang N, Hu H, Li J, Ouyang F, Wang J, Zhao Y, Ma Z, Meng C, Li Z, Yu S, Jing J, Zheng M. Centripetal migration and prolonged retention of microglia promotes spinal cord injury repair. J Neuroinflammation 2025; 22:77. [PMID: 40075472 PMCID: PMC11905688 DOI: 10.1186/s12974-025-03411-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Recent studies have confirmed the critical role of neonatal microglia in wound healing and axonal regeneration following spinal cord injury (SCI). However, the limited migration of microglia to the center of adult lesion may significantly impede their potential benefits. METHODS We established a model of microglial centripetal migration and prolonged retention in C57BL/6J and transgenic mice by injecting exogenous C-X3-C motif chemokine ligand 1 (CX3CL1) and macrophage colony-stimulating factor (M-CSF) directly into the lesion site post-SCI. Wound healing and axonal preservation/regrowth was assessed anatomically, and kinematics analysis was conducted to determine the recovery of locomotor function. RESULTS We identified decreased expression and perilesional distribution of CX3CL1 as the primary reason for the limited centripetal migration of microglia. In situ injection of CX3CL1 into the lesion core promoted microglial centripetal migration, but alone did not improve functional recovery. Nevertheless, a combinational administration of CX3CL1 and M-CSF fostered both centripetal migration and prolonged retention of microglia, thereby effectively displacing blood-derived macrophage infiltration and optimizing wound healing and axonal preservation/regrowth after SCI. Notably, the beneficial effects of CX3CL1 and M-CSF co-administration were specifically blocked in C-X3-C motif chemokine receptor 1 (CX3CR1)-deficient mice. These phenomena may be related to the increase in spleen tyrosine kinase (SYK) levels, which boosts centripetal microglial phagocytosis. CONCLUSION Our study uncovers the criticality of microglial location and abundance in orchestrating SCI repair, highlighting centripetal microglial dynamics as valuable targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jianan Ye
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fangli Shan
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Xinzhong Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Chao Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ningyuan Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hao Hu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianjian Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fangru Ouyang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jingwen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuanzhe Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhida Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Congpeng Meng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ziyu Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Shuisheng Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Juehua Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Meige Zheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
37
|
Schaible P, Henschel J, Erny D. How the gut microbiota impacts neurodegenerative diseases by modulating CNS immune cells. J Neuroinflammation 2025; 22:60. [PMID: 40033338 PMCID: PMC11877772 DOI: 10.1186/s12974-025-03371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. Amyloid-β (Aβ) accumulation and neurofibrillary tangles are two key histological features resulting in progressive and irreversible neuronal loss and cognitive decline. The macrophages of the central nervous system (CNS) belong to the innate immune system and comprise parenchymal microglia and CNS-associated macrophages (CAMs) at the CNS interfaces (leptomeninges, perivascular space and choroid plexus). Microglia and CAMs have received attention as they may play a key role in disease onset and progression e. g., by clearing amyloid beta (Aβ) through phagocytosis. Genome-wide association studies (GWAS) have revealed that human microglia and CAMs express numerous risk genes for AD, further highlighting their potentially critical role in AD pathogenesis. Microglia and CAMs are tightly controlled by environmental factors, such as the host microbiota. Notably, it was further reported that the composition of the gut microbiota differed between AD patients and healthy individuals. Hence, emerging studies have analyzed the impact of gut bacteria in different preclinical mouse models for AD as well as in clinical studies, potentially enabling promising new therapeutic options.
Collapse
Affiliation(s)
- Philipp Schaible
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Julia Henschel
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Erny
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| |
Collapse
|
38
|
Zhang Y, Liu Y, Luo S, Liang H, Guo C, Du Y, Li H, Wang L, Wang X, Tang C, Zhou Y. An adoptive cell therapy with TREM2-overexpressing macrophages mitigates the transition from acute kidney injury to chronic kidney disease. Clin Transl Med 2025; 15:e70252. [PMID: 40000418 PMCID: PMC11859120 DOI: 10.1002/ctm2.70252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Macrophages have been shown to contribute to renal injury and fibrosis as well as repair. Recently, Triggering Receptor Expressed on Myeloid Cells 2 (TREM2)-positive macrophages have been shown to play important roles in regulating tissue inflammation and repair. However, it remains unclear whether they can mitigate the transition from acute kidney injury to chronic kidney disease (the AKI-CKD transition). METHODS The AKI-CKD transition was generated by unilateral ischaemia-reperfusion injury (UIRI) in wild-type (WT) and Trem2 knockout mice. F4/80 magnetic beads were used to isolate renal macrophages. Flow cytometry was used to determine the levels of TREM2 and CD11b levels. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blotting and histological staining were performed to determine the expression of cytokines and fibrotic markers. RNA-seq was used to investigate transcriptomic changes between WT and Trem2 knockout bone marrow-derived macrophages (BMDMs). TREM2-overexpressing macrophages were generated using lentivirus and transferred intravenously to UIRI mice. RESULTS TREM2 macrophages exhibited a strong renal protective effect on the AKI-CKD transition. Genetic deletion of Trem2 resulted in increased renal inflammation and exacerbated renal injury and fibrosis in UIRI mice. Interestingly, we found that hypoxia could increase TREM2 expression in macrophages via HIF-1α. Upregulated TREM2 expression enhanced macrophage phagocytosis and suppressed the expression of pro-inflammatory cytokines, resulting in lower levels of apoptosis and fibrosis in tubular epithelial cells. Using RNA-seq analysis, we showed that the regulatory effects of TREM2 were orchestrated by the PI3K-AKT pathway. Pharmacological regulation of the PI3K-AKT pathway could modulate the macrophage-mediated inflammation and phagocytosis. In addition, an adoptive cell therapy using TREM2-overexpressing macrophages effectively reduced the immune cell infiltration, renal injury and fibrosis in UIRI mice. CONCLUSION Our study not only provides valuable mechanistic insights into the role of Trem2 in the AKI-CKD transition but also offers a new avenue for TREM2-overexpressing macrophage-based adoptive cell therapy to treat kidney diseases. KEY POINTS TREM2 knockout worsens kidney injury and accelerates AKI-CKD transition. TREM2 is upregulated by hypoxia via HIF1α in AKI-CKD transition. An adoptive cell therapy using TREM2-overexpressing macrophages reduces kidney inflammation and fibrosis.
Collapse
Affiliation(s)
- Yating Zhang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yu Liu
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Siweier Luo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hanzhi Liang
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Chipeng Guo
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yufei Du
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hongyu Li
- Department of Nephrology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Le Wang
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaohua Wang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Chun Tang
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yiming Zhou
- Basic and Translational Medical Research Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
39
|
Fang M, Zhou Y, He K, Lu Y, Tao F, Huang H. Glucose Metabolic Reprogramming in Microglia: Implications for Neurodegenerative Diseases and Targeted Therapy. Mol Neurobiol 2025:10.1007/s12035-025-04775-y. [PMID: 39987285 DOI: 10.1007/s12035-025-04775-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
As intrinsic immune cells in the central nervous system, microglia play a crucial role in maintaining brain homeostasis. Microglia can transition from homeostasis to various responsive states in reaction to different external stimuli, undergoing corresponding alterations in glucose metabolism. In neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS), microglial glucose metabolic reprogramming is widespread. This reprogramming leads to changes in microglial function, exacerbating neuroinflammation and the accumulation of pathological products, thereby driving the progression of neurodegeneration. This review summarizes the specific alterations in glucose metabolism within microglia in AD, PD, ALS, and MS, as well as the corresponding treatments aimed at reprogramming glucose metabolism. Compounds that inhibit key glycolytic enzymes like hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), or activate regulators of energy metabolism such as AMP-activated protein kinase (AMPK), have shown significant potential in the treatment of various neurodegenerative diseases. However, current research faces numerous challenges, including side effects and blood-brain barrier (BBB) penetration of compounds. Screening relevant drugs from natural products, especially flavonoids, is a reliable approach. On the one hand, longtime herbal medical practices provide a certain degree of assurance regarding clinical safety, and their chemical properties contribute to effective BBB permeability. On the other hand, the concurrent anti-tumor and anti-neuroinflammatory activities of flavonoids suggest that regulation of glucose metabolism reprogramming might be a potential common mechanism of action. Notably, considering the dynamic nature of microglial metabolism, there is an urgent need to develop technologies for real-time monitoring of glucose metabolism processes, which would significantly advance research in this field.
Collapse
Affiliation(s)
- Mengqi Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Keren He
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yangyuxiao Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Fangfang Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Hong Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
40
|
You T, Yang Y, A L, Cheng X, Lin X, Liang Q, Ge L, Xie J, Chen S, Liu N, He J, Xu H, Ma X. IFNγ preconditioning improves neuroprotection of MSC-derived vesicles on injured retinal ganglion cells by suppressing microglia activation via miRNA-dependent ribosome activity. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:87-111. [PMID: 40206798 PMCID: PMC11977360 DOI: 10.20517/evcna.2024.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/19/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Aim: Microglial activation plays a pivotal role in the pathogenesis of retinal ganglion cell (RGC) degeneration resulting from optic nerve crush (ONC). Small extracellular vesicles (sEVs) secreted by mesenchymal stem cells (MSCs) have the potential to prevent retinal degeneration by modulating microglial activation. In this study, we elucidated the specific effects of sEVs derived from IFN-γ-primed MSCs on the phenotypic transition of microglia and the associated pathways in ONC mice. Methods: The ONC mice model was established and administered intravitreal injection with the sEVs derived from native MSCs (native sEVs) and the sEVs derived from MSCs primed with IFN-γ (IFNγ-sEVs). Their respective effects on the survival of the retinal ganglion cells (RGCs) and the transition of microglia phenotypes were determined through visual function testing and immunohistochemical staining. Combined with mRNA seq and microRNA seq techniques, we elucidated the mechanism of modulation of microglia phenotypic transformation by sEVs derived from MSCs primed by IFNγ. Results: It demonstrated that IFNγ-sEVs exhibited superior protective effects against RGC loss and reduced inflammatory responses in the ONC retina compared to native sEVs. Both types of sEVs promoted microglia activation to disease-associated microglia (DAM) phenotype, while IFNγ-sEVs especially suppressed interferon-responsive microglia (IRM) activation during RGCs degeneration. Subsequent miRNA sequencing suggested that miR-423-5p, which exhibited the most significant differential expression between the two sEVs types and elevated expression in IFNγ-sEVs, inhibited the expression of IRM and ribosomal genes. Conclusion: These findings suggest that IFN-γ-preconditioned MSCs may enhance sEVs of neuroprotection on RGCs by suppressing IRM activation through the secretion of sEVs containing specific microRNAs in ONC mice.
Collapse
Affiliation(s)
- Tianjing You
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Yuanxing Yang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Luodan A
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Authors contributed equally
| | - Xuan Cheng
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xi Lin
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Qingle Liang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Jing Xie
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Siyu Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Na Liu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Juncai He
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
- Department of Ophthalmology, The 920 Hospital of PLA Joint Logistics Support Force, Kunming 650032, Yunnan, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Amy Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration and Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing 400038, China
| | - Xiang Ma
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian 116014, Liaoning, China
| |
Collapse
|
41
|
Medd MM, Yon JE, Dong H. RhoA/ROCK/GSK3β Signaling: A Keystone in Understanding Alzheimer's Disease. Curr Issues Mol Biol 2025; 47:124. [PMID: 39996845 PMCID: PMC11854763 DOI: 10.3390/cimb47020124] [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: 01/19/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline and loss of neuronal integrity. Emerging evidence suggests that RhoA, Rho-associated coiled-coil kinase (ROCK), and their downstream effector molecule glycogen synthase 3β (GSK3β) interact within a complex signaling pathway (RhoA/ROCK/GSK3β) that plays a crucial role in the pathogenesis of AD. RhoA, a small GTPase, along with its downstream effector, ROCK, regulates various cellular processes, including actin cytoskeleton dynamics, apoptosis, and synaptic plasticity. GSK3β, a serine/threonine kinase, plays a key role in neuronal function and AD pathology, including the regulation of tau phosphorylation and amyloid-beta cleavage. Overactive GSK3β has been closely linked to tau hyperphosphorylation, neurodegeneration, and the progression of AD. Thus, GSK3β has been considered as a promising therapeutic target for treating AD and mitigating cognitive impairment. However, clinical trials of GSK3β in AD have faced considerable challenges due to the complexity of the specific neuronal inhibition of GSK3β. In this review, we summarize the literature regarding the relationship of RhoA/ROCK and GSK3β signaling pathways in AD pathogenesis. We further discuss recent findings of the sTREM2-transgelin-2 (TG2) axis as a potential mediator of this complex pathway and provide our review on a novel targeting strategy for AD.
Collapse
Affiliation(s)
- Milan M. Medd
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.M.M.); (J.E.Y.)
| | - Jayden E. Yon
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.M.M.); (J.E.Y.)
| | - Hongxin Dong
- Stephen M. Stahl Center for Psychiatric Neuroscience, Departments of Psychiatry & Behavioral Sciences and Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
42
|
Fu J, Wang R, He J, Liu X, Wang X, Yao J, Liu Y, Ran C, Ye Q, He Y. Pathogenesis and therapeutic applications of microglia receptors in Alzheimer's disease. Front Immunol 2025; 16:1508023. [PMID: 40028337 PMCID: PMC11867950 DOI: 10.3389/fimmu.2025.1508023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Microglia, the resident immune cells of the central nervous system, continuously monitor the brain's microenvironment through their array of specific receptors. Once brain function is altered, microglia are recruited to specific sites to perform their immune functions, including phagocytosis of misfolded proteins, cellular debris, and apoptotic cells to maintain homeostasis. When toxic substances are overproduced, microglia are over-activated to produce large amounts of pro-inflammatory cytokines, which induce chronic inflammatory responses and lead to neurotoxicity. Additionally, microglia can also monitor and protect neuronal function through microglia-neuron crosstalk. Microglia receptors are important mediators for microglia to receive external stimuli, regulate the functional state of microglia, and transmit signals between cells. In this paper, we first review the role of microglia-expressed receptors in the pathogenesis and treatment of Alzheimer's disease; moreover, we emphasize the complexity of targeting microglia for therapeutic interventions in neurodegenerative disorders to inform the discovery of new biomarkers and the development of innovative therapeutics.
Collapse
Affiliation(s)
- Jiao Fu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - RuoXuan Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JiHui He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XiaoJing Liu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JuMing Yao
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - ChongZhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - QingSong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
- Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Beltran-Velasco AI. Brain Glycogen-Its Metabolic Role in Neuronal Health and Neurological Disorders-An Extensive Narrative Review. Metabolites 2025; 15:128. [PMID: 39997753 PMCID: PMC11857135 DOI: 10.3390/metabo15020128] [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: 01/06/2025] [Revised: 01/29/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Brain glycogen is imperative for neuronal health, as it supports energy demands and metabolic processes. This review examines the pathways involved in glycogen storage and utilization in the central nervous system, emphasizing their role in both physiology and pathology. It explores how alterations in glycogen metabolism contribute to neurological disorders, including neurodegenerative diseases, epilepsy, and metabolic conditions while highlighting the bidirectional interaction between neurons and glia in maintaining brain homeostasis. Methods: A comprehensive search of articles published between 2015 and 2025 was conducted using the following databases: ScienceDirect, Scopus, Wiley, Web of Science, Medline, and PubMed. The selection of relevant studies was based on their focus on brain glycogen metabolism and its role in neurological conditions, with studies that did not meet the inclusion criteria being excluded. Results: The metabolic processes of brain glycogen are subject to rigorous regulation by astrocyte-neuron interactions, thereby ensuring metabolic homeostasis and energy availability. The dysregulation of glycogen storage and mobilization has been implicated in the development of synaptic dysfunction, excitotoxicity, and neurodegeneration in a variety of disorders. For instance, aberrant glycogen accumulation in diseases such as Lafora disease has been associated with severe neurodegeneration, while impaired glycogen mobilization has been shown to exacerbate energy deficits in Alzheimer's and epilepsy. Conclusions: Targeting brain glycogen metabolism represents a promising approach for therapeutic intervention in neurological disorders. However, the translation of these strategies to human models remains challenging, particularly with regard to the long-term safety and specificity of glycogen-targeted therapies.
Collapse
|
44
|
Zhao M, Jankovic D, Link VM, Souza COS, Hornick KM, Oyesola O, Belkaid Y, Lack J, Loke P. Genetic variation in IL-4 activated tissue resident macrophages determines strain-specific synergistic responses to LPS epigenetically. Nat Commun 2025; 16:1030. [PMID: 39863579 PMCID: PMC11762786 DOI: 10.1038/s41467-025-56379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
How macrophages in the tissue environment integrate multiple stimuli depends on the genetic background of the host, but this is still poorly understood. We investigate IL-4 activation of male C57BL/6 and BALB/c strain specific in vivo tissue-resident macrophages (TRMs) from the peritoneal cavity. C57BL/6 TRMs are more transcriptionally responsive to IL-4 stimulation, with induced genes associated with more super enhancers, induced enhancers, and topologically associating domains (TAD) boundaries. IL-4-directed epigenomic remodeling reveals C57BL/6 specific enrichment of NF-κB, IRF, and STAT motifs. Additionally, IL-4-activated C57BL/6 TRMs demonstrate an augmented synergistic response upon in vitro lipopolysaccharide (LPS) exposure, despite naïve BALB/c TRMs displaying a more robust transcriptional response to LPS. Single-cell RNA sequencing (scRNA-seq) analysis of mixed bone marrow chimeras indicates that transcriptional differences and synergy are cell intrinsic within the same tissue environment. Hence, genetic variation alters IL-4-induced cell intrinsic epigenetic reprogramming resulting in strain specific synergistic responses to LPS exposure.
Collapse
Affiliation(s)
- Mingming Zhao
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dragana Jankovic
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Camila Oliveira Silva Souza
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine M Hornick
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Oyebola Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Png Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
45
|
Lee E, Chang Y. Modulating Neuroinflammation as a Prospective Therapeutic Target in Alzheimer's Disease. Cells 2025; 14:168. [PMID: 39936960 PMCID: PMC11817173 DOI: 10.3390/cells14030168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
The recent approval of lecanemab highlights that the amyloid beta (Aβ) protein is an important pathological target in Alzheimer's disease (AD) and further emphasizes the significance of neuroinflammatory pathways in regulating Aβ accumulation. Indeed, Aβ accumulation triggers microglia activation, which are key mediators in neuroinflammation. The inflammatory responses in this process can lead to neuronal damage and functional decline. Microglia secrete proinflammatory cytokines that accelerate neuronal death and release anti-inflammatory cytokines and growth factors contributing to neuronal recovery and protection. Thus, microglia play a dual role in neurodegeneration and neuroprotection, complicating their function in AD. Therefore, elucidating the complex interactions between Aβ protein, microglia, and neuroinflammation is essential for developing new strategies for treating AD. This review investigates the receptors and pathways involved in activating microglia and aims to enhance understanding of how these processes impact neuroinflammation in AD, as well as how they can be regulated. This review also analyzed studies reported in the existing literature and ongoing clinical trials. Overall, these studies will contribute to understanding the regulatory mechanisms of neuroinflammation and developing new therapies that can slow the pathological progression of AD.
Collapse
Affiliation(s)
- Eunshil Lee
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
| | - Yongmin Chang
- Institute of Biomedical Engineering Research, Kyungpook National University, Daegu 41944, Republic of Korea;
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Radiology, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| |
Collapse
|
46
|
Wang F, Wang J, Luo N, Luo Y, Gao Z, Cui Y, Jiang M, Shen Z, Xiao J, Xue P, Wang X, Yu S, Wu Q, Zhao X. Exposure to nano-polystyrene during pregnancy leads to Alzheimer's disease-related pathological changes in adult offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117736. [PMID: 39818142 DOI: 10.1016/j.ecoenv.2025.117736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/03/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Nanoplastics are common environmental pollutants. As of now, research has yet to explore how exposure to nanomaterials during gestation might influence the risk of developing Alzheimer's disease (AD) in offspring. Throughout the research, we assessed the AD pathology in adult offspring of mice prenatal 80 nm polystyrene nanoparticles (PS-NPs) exposure. In contrast with the control group, prenatal PS-NPs exposure obviously decreased brain tissue weight and the organ coefficient (brain weight/body weight) in adult male mice, but it only led to changes in the low-dose group of female mice. Histological examination of the adult offspring brains revealed alterations following exposure to PS-NPs during gestation. Specifically, there was a substantial reduction in neuron cells, significant changes in the number of Nissl bodies, noticeable loss of cell nucleus, and increased presence of neurofibrillary tangles in adult offspring mice exposed to PS-NPs during gestation. Furthermore, the phosphorylation levels of tau proteins at ser396 and ser199 were dramatically enhanced in the PS-NPs exposed group. Furthermore, the expression of Aβ protein was markedly increased, consistent with typical AD pathological features. Our findings suggest that being exposed to PS-NPs during pregnancy substantially raises the risk of AD in offspring.
Collapse
Affiliation(s)
- Fengxu Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Jin Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Na Luo
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yonghua Luo
- Nantong Fourth People's Hospital, Nantong, China
| | - Zhengnan Gao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Ye Cui
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Mengna Jiang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Zhaoping Shen
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Jing Xiao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Xiaoke Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| | - Qiyun Wu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
47
|
Gharibani P, Abramson E, Shanmukha S, Smith MD, Godfrey WH, Lee JJ, Hu J, Baydyuk M, Dorion MF, Deng X, Guo Y, Calle AJ, A Hwang S, Huang JK, Calabresi PA, Kornberg MD, Kim PM. The protein kinase C modulator bryostatin-1 therapeutically targets microglia to attenuate neuroinflammation and promote remyelination. Sci Transl Med 2025; 17:eadk3434. [PMID: 39772770 DOI: 10.1126/scitranslmed.adk3434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In multiple sclerosis (MS), microglia and macrophages within the central nervous system (CNS) play an important role in determining the balance among demyelination, neurodegeneration, and myelin repair. Phagocytic and regenerative functions of these CNS innate immune cells support remyelination, whereas chronic and maladaptive inflammatory activation promotes lesion expansion and disability, particularly in the progressive forms of MS. No currently approved drugs convincingly target microglia and macrophages within the CNS, contributing to the lack of therapies aimed at promoting remyelination and slowing disease progression for individuals with MS. Here, we found that the protein kinase C (PKC)-modulating drug bryostatin-1 (bryo-1), a CNS-penetrant compound with an established human safety profile, shifts the transcriptional programs of microglia and CNS-associated macrophages from a proinflammatory phenotype to a regenerative phenotype in vitro and in vivo. Treatment of microglia with bryo-1 stimulated scavenger pathways, phagocytosis, and secretion of factors that prevented the activation of neuroinflammatory reactive astrocytes while also promoting neuroaxonal health and oligodendrocyte differentiation. In line with these findings, systemic treatment of mice with bryo-1 augmented remyelination after a focal demyelinating injury. Our results demonstrate the potential of bryo-1 and possibly a wider class of PKC modulators as myelin-regenerative and supportive agents in MS and other neurologic diseases.
Collapse
Affiliation(s)
- Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Efrat Abramson
- Interdepartmental Neuroscience Program, Yale University School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wesley H Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Judy J Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jingwen Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew J Calle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Soonmyung A Hwang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
48
|
Shahidehpour RK, Nelson PT, Katsumata Y, Bachstetter AD. Exploring the link between dystrophic microglia and the spread of Alzheimer's neuropathology. Brain 2025; 148:89-101. [PMID: 39101580 PMCID: PMC11706277 DOI: 10.1093/brain/awae258] [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: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/15/2024] [Indexed: 08/06/2024] Open
Abstract
Genetics and other data modalities indicate that microglia play a critical role in Alzheimer's disease progression, but details of the disease-driving influence of microglia are poorly understood. Microglial cells can be parsed into subtypes based on their histological appearance. One subtype of microglia, termed dystrophic microglia, is characterized structurally by fragmented processes and cytoplasmic decay, and their presence has been associated with ageing and neurodegeneration. Recent studies suggest that the interaction between tau proteins and amyloid-β might induce dystrophic changes in microglia, potentially linking amyloid-β and tau pathologies to their effects on these microglia. We developed a study of human brains to test the hypothesis that dystrophic microglia are involved in Alzheimer's disease progression. We speculated that if their presence is unique to Alzheimer's disease neuropathological change, they would be substantially more common in Alzheimer's disease neuropathological change than in neurodegenerative diseases characterized by other proteinopathies, e.g. α-synuclein or transactive response (TAR) DNA-binding protein 43 kDa (TDP-43) pathology. Our analyses used histologically stained sections from five human brain regions of 64 individuals across six disease states, from healthy controls to advanced Alzheimer's disease stages, including comparative conditions such as Lewy body disease and limbic-predominant age-related TDP-43 encephalopathy neuropathological change. Using stereological sampling and digital pathology, we assessed populations of ramified, hypertrophic and dystrophic microglia. We found a significant increase in dystrophic microglia in areas affected early by Alzheimer's disease neuropathological change, suggesting a disease-specific role in neuropathology. Mediation analysis and structural equation modelling suggest that dystrophic microglia might impact the regional spread of Alzheimer's disease neuropathological change. In the mediation model, tau was found to be the initiating factor leading to the development of dystrophic microglia, which was then associated with the spread of amyloid-β and tau. These results suggest that a loss of the protective role of microglia could contribute to the spread of Alzheimer's disease neuropathological change and indicate that further research into preserving microglial function might be warranted.
Collapse
Affiliation(s)
- Ryan K Shahidehpour
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Department of Pathology and Laboratory Medicine, Division of Neuropathology, University of Kentucky, Lexington, KY 40536, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
49
|
Pianeta R, Deosthale P, Sanz N, Kohler R, Okpara C, Arnett M, Asad I, Rogers A, Gerbig M, Essex A, Liu Z, Wallace JM, Plotkin LI. Sex hormone deficiency in male and female mice expressing the Alzheimer's disease-associated risk-factor TREM2 R47H variant impacts the musculoskeletal system in a sex- and genotype-dependent manner. JBMR Plus 2025; 9:ziae144. [PMID: 39677924 PMCID: PMC11646090 DOI: 10.1093/jbmrpl/ziae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/25/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
The R47H variant of the triggering receptor expressed on myeloid cells 2 (TREM2) is a risk factor for Alzheimer's disease in humans and leads to lower bone mass accrual in female but not male 12-mo-old mice. To determine whether, as with aging, gonadectomy results in sex-specific musculoskeletal effects, gonad removal or SHAM surgery was performed in 4-mo-old TREM2R47H/+ mice and WT male and female littermates (n = 10-12/group), with sexes analyzed separately. Body weight was lower in males, but higher in females after gonadectomy, independently of their genotype. Gonadectomy also leads to decreased BMD in males at all sites and in the whole body (total) and spine in female mice for both genotypes. Total and femur BMD was lower in gonadectomized male mice 6-wk post-surgery, independently of the genotype. On the other hand, BMD was only lower in ovariectomized WT but not TREM2R47H/+ mice in all sites measured at this time point. Bone formation and resorption marker levels were not affected by orchiectomy, whereas CTX was higher 3 wk after surgery and P1NP showed a tendency toward lower values at the 6-wk time point only in ovariectomized WT mice. Micro-CT analyses showed no differences resulting from gonadectomy in structural parameters in femoral cortical bone for either sex, but lower tissue mineral density in males of either genotype 6-wk post-surgery. Nevertheless, biomechanical properties were overall lower in gonadectomized males of either genotype, and only for WT ovariectomized mice. Distal femur cancellous bone structure was also affected by gonadectomy in a genotype- and sex-dependent manner, with genotype-independent changes in males, and only in WT female mice. Thus, expression of the TREM2 R47H variant minimally alters the impact of gonadectomy in the musculoskeletal system in males, whereas it partially ameliorates the consequences of ovariectomy in female mice.
Collapse
Affiliation(s)
- Roquelina Pianeta
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
| | - Padmini Deosthale
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
| | - Natasha Sanz
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Bone Biology Laboratory, School of Medicine, Rosario National University, Rosario 2000, Argentina
- National Council of Scientific and Technical Research (CONICET), Buenos Aires 9100, Argentina
| | - Rachel Kohler
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Chiebuka Okpara
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Matthew Arnett
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Iqra Asad
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Amber Rogers
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Madison Gerbig
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Alyson Essex
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Ziyue Liu
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Joseph M Wallace
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
- Department of Biomechanical Engineering, Indiana University—Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
| |
Collapse
|
50
|
Zhang Z, Wu M, Yao L, Zhou W, Liu X, Chen Z, Hua P, Xu L, Lv L, Liu C, Huang C, Chen S, Huang Z, Huang Y, He J, Chen T, Wang J, Yuan W, Liu Z, Chen Y. Trem2/Tyrobp Signaling Protects Against Aortic Dissection and Rupture by Inhibiting Macrophage Activation in Mice. Arterioscler Thromb Vasc Biol 2025; 45:119-135. [PMID: 39508103 DOI: 10.1161/atvbaha.124.321429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND The development of aortic dissection (AD) is closely associated with inflammation. The Trem2 (triggering receptor expressed on myeloid cells 2)/Tyrobp (TYRO protein tyrosine kinase-binding protein) signaling pathway critically regulates innate immunity and has emerged as an important target in cardiovascular diseases; however, its role in AD remains unclear. METHODS Transcriptome data from human and mouse ADs were used to perform differentially expressed gene-based protein-protein interaction network analyses. Tyrobp knockout (Tyrobp-/-), myeloid cell-specific Tyrobp-/- (Tyrobpfl/fl Lyz2cre), and Trem2 knockout (Trem2-/-) mice were given β-aminopropionitrile monofumarate in drinking water to induce AD. To dissect the role of macrophages in Tyrobp deficiency-mediated AD progression, macrophages were depleted using clodronate liposomes. Bulk and single-cell RNA sequencing, immunofluorescence staining, and quantitative real-time polymerase chain reaction were performed to assess inflammation and the underlying mechanisms of Tyrobp in AD. RESULTS Network analysis identified Tyrobp as a hub gene of AD, with elevated levels observed in both human and mouse ADs. Global deletion and myeloid cell-specific deficiency of Tyrobp in mice significantly increased AD incidence and exacerbated extracellular matrix degradation and macrophage infiltration within the aortic wall. Macrophage depletion mitigated the adverse effects of Tyrobp deficiency on AD progression. Additionally, Tyrobp deficiency enhanced TLR (Toll-like receptor)-4 signaling and macrophage activation, which were abrogated by TLR4 inhibitors. Furthermore, deletion of the Tyrobp-associated receptor Trem2 significantly aggravated mouse AD development, whereas Trem2 agonist treatment conferred protection against AD. CONCLUSIONS Our findings suggest a novel role for the Trem2/Tyrobp axis in AD development in mice. Enhancement of Trem2/Tyrobp signaling may represent a promising strategy for the prevention and treatment of AD. Future studies to clarify the role of Trem2/Tyrobp in human AD are warranted.
Collapse
Affiliation(s)
- Zenghui Zhang
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China (Z.Z.)
| | - Maoxiong Wu
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Yao
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weibin Zhou
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao Liu
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhiteng Chen
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Hua
- Department of Cardio-Vascular Surgery (P.H.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Leibo Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Biliary-Pancreatic Surgery (L.X.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Lv
- Department of Cardiac and Vascular Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Yunnan, China (L.L.)
| | - Chiyu Liu
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chunling Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Sixu Chen
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaoqi Huang
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuna Huang
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiaqi He
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tingfeng Chen
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingfeng Wang
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Woliang Yuan
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhaoyu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yangxin Chen
- Department of Cardiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation (Z.Z., M.W., L.Y., W.Z., X.L., Z.C., C.L., C.H., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Z.L., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Molecular Mechanism and Translation in Major Cardiovascular Disease (Z.Z., M.W., W.Z., X.L., Z.C., C.L., S.C., Z.H., Y.H., J.H., T.C., J.W., W.Y., Y.C.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|