1
|
Frolov A, Huang H, Schütz D, Köhne M, Blank-Stein N, Osei-Sarpong C, Büttner M, Elmzzahi T, Khundadze M, Zahid M, Reuter M, Becker M, De Domenico E, Bonaguro L, Kallies A, Morrison H, Hübner CA, Händler K, Stumm R, Mass E, Beyer MD. Microglia and CD8+ T cell activation precede neuronal loss in a murine model of spastic paraplegia 15. J Exp Med 2025; 222:e20232357. [PMID: 40266307 PMCID: PMC12017274 DOI: 10.1084/jem.20232357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
In central nervous system (CNS) diseases characterized by late-onset neurodegeneration, the interplay between innate and adaptive immune responses remains poorly understood. This knowledge gap is exacerbated by the prolonged protracted disease course as it complicates the delineation of brain-resident and infiltrating cells. Here, we conducted comprehensive profiling of innate and adaptive immune cells in a murine model of spastic paraplegia 15 (SPG15), a complicated form of hereditary spastic paraplegia. Using fate-mapping of bone marrow-derived cells, we identified microgliosis accompanied by infiltration and local expansion of T cells in the CNS of Spg15-/- mice. Single-cell analysis revealed an expansion of disease-associated microglia (DAM) and effector CD8+ T cells prior to neuronal loss. Analysis of potential cell-cell communication pathways suggested bidirectional interactions between DAM and effector CD8+ T cells, potentially contributing to disease progression in Spg15-/- mice. In summary, we identified a shift in microglial phenotypes associated with the recruitment and expansion of T cells as a new characteristic of Spg15-driven neuropathology.
Collapse
Affiliation(s)
- Aleksej Frolov
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Hao Huang
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
| | - Maren Köhne
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Nelli Blank-Stein
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Collins Osei-Sarpong
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Institute of Experimental Pathology, Centre of Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Maren Büttner
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Tarek Elmzzahi
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Mukhran Khundadze
- Institute of Human Genetics, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
- Center for Rare Diseases, University Hospital Jena, Friedrich-Schiller-University, Jena, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marina Zahid
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Michael Reuter
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Matthias Becker
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Modular High-Performance Computing and Artificial Intelligence, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Elena De Domenico
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Axel Kallies
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany
| | - Christian A. Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
- Center for Rare Diseases, University Hospital Jena, Friedrich-Schiller-University, Jena, Germany
| | - Kristian Händler
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
- Institute of Human Genetics, Universitätsklinikum Schleswig-Holstein, University of Lübeck and University of Kiel, Lübeck, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University Jena, Jena, Germany
| | - Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Marc D. Beyer
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| |
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
|
González-Velasco O, Simon M, Yilmaz R, Parlato R, Weishaupt J, Imbusch C, Brors B. Identifying similar populations across independent single cell studies without data integration. NAR Genom Bioinform 2025; 7:lqaf042. [PMID: 40276039 PMCID: PMC12019640 DOI: 10.1093/nargab/lqaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Supervised and unsupervised methods have emerged to address the complexity of single cell data analysis in the context of large pools of independent studies. Here, we present ClusterFoldSimilarity (CFS), a novel statistical method design to quantify the similarity between cell groups across any number of independent datasets, without the need for data correction or integration. By bypassing these processes, CFS avoids the introduction of artifacts and loss of information, offering a simple, efficient, and scalable solution. This method match groups of cells that exhibit conserved phenotypes across datasets, including different tissues and species, and in a multimodal scenario, including single-cell RNA-Seq, ATAC-Seq, single-cell proteomics, or, more broadly, data exhibiting differential abundance effects among groups of cells. Additionally, CFS performs feature selection, obtaining cross-dataset markers of the similar phenotypes observed, providing an inherent interpretability of relationships between cell populations. To showcase the effectiveness of our methodology, we generated single-nuclei RNA-Seq data from the motor cortex and spinal cord of adult mice. By using CFS, we identified three distinct sub-populations of astrocytes conserved on both tissues. CFS includes various visualization methods for the interpretation of the similarity scores and similar cell populations.
Collapse
Affiliation(s)
- Oscar González-Velasco
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Malte Simon
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Leibniz Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Rüstem Yilmaz
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Rosanna Parlato
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Jochen Weishaupt
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 68167 Mannheim, Germany
| | - Charles D Imbusch
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Immunology, University Medical Center Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Benedikt Brors
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Medical Faculty Heidelberg and Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
4
|
Hou J, Hess JL, Zhang C, van Rooij JGJ, Hearn GC, Fan CC, Faraone SV, Fennema-Notestine C, Lin SJ, Escott-Price V, Seshadri S, Holmans P, Tsuang MT, Kremen WS, Gaiteri C, Glatt SJ. Meta-Analysis of Transcriptomic Studies of Blood and Six Brain Regions Identifies a Consensus of 15 Cross-Tissue Mechanisms in Alzheimer's Disease and Suggests an Origin of Cross-Study Heterogeneity. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33019. [PMID: 39679839 PMCID: PMC12048288 DOI: 10.1002/ajmg.b.33019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024]
Abstract
The comprehensive genome-wide nature of transcriptome studies in Alzheimer's disease (AD) should provide a reliable description of disease molecular states. However, the genes and molecular systems nominated by transcriptomic studies do not always overlap. Even when results do align, it is not clear if those observations represent true consensus across many studies. A couple of sources of variation have been proposed to explain this variability, including tissue-of-origin and cohort type, but its basis remains uncertain. To address this variability and extract reliable results, we utilized all publicly available blood or brain transcriptomic datasets of AD, comprised of 24 brain studies with 4007 samples from six different brain regions, and eight blood studies with 1566 samples. We identified a consensus of AD-associated genes across brain regions and AD-associated gene-sets across blood and brain, generalizable machine learning and linear scoring classifiers, and significant contributors to biological diversity in AD datasets. While AD-associated genes did not significantly overlap between blood and brain, our findings highlighted 15 dysregulated processes shared across blood and brain in AD. The top five most significantly dysregulated processes were DNA replication, metabolism of proteins, protein localization, cell cycle, and programmed cell death. Conversely, addressing the discord across studies, we found that large-scale gene co-regulation patterns can account for a significant fraction of variability in AD datasets. Overall, this study ranked and characterized a compilation of genes and molecular systems consistently identified across a large assembly of AD transcriptome studies in blood and brain, providing potential candidate biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jiahui Hou
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jonathan L Hess
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chunling Zhang
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jeroen G J van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gentry C Hearn
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Chun Chieh Fan
- Department of Cognitive Science, University of California San Diego, La Jolla, California, USA
| | - Stephen V Faraone
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Shu-Ju Lin
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Valentina Escott-Price
- Dementia Research Institute, School of Medicine, Cardiff University, Cardiff, UK
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Sudha Seshadri
- Department of Neurology, School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Peter Holmans
- Division of Psychological Medicine and Clinical Neurology and Medical Research Council (MRC) Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Chris Gaiteri
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Stephen J Glatt
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Public Health and Preventive Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
5
|
Murphy KB, Ye Y, Tsalenchuk M, Nott A, Marzi SJ. CHAS infers cell type-specific signatures in bulk brain histone acetylation studies of neurological and psychiatric disorders. CELL REPORTS METHODS 2025; 5:101032. [PMID: 40300607 DOI: 10.1016/j.crmeth.2025.101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/07/2025] [Accepted: 04/04/2025] [Indexed: 05/01/2025]
Abstract
Epigenomic profiling of the brain has largely been done on bulk tissues, limiting our understanding of cell type-specific epigenetic changes in disease states. Here, we introduce cell type-specific histone acetylation score (CHAS), a computational tool for inferring cell type-specific signatures in bulk brain H3K27ac profiles. We applied CHAS to >300 H3K27ac chromatin immunoprecipitation sequencing samples from studies of Alzheimer's disease, Parkinson's disease, autism spectrum disorder, schizophrenia, and bipolar disorder in bulk postmortem brain tissue. In addition to recapitulating known disease-associated shifts in cellular proportions, we identified cell type-specific biological insights into brain-disorder-associated regulatory variation. In most cases, genetic risk and epigenetic dysregulation targeted different cell types, suggesting independent mechanisms. For instance, genetic risk of Alzheimer's disease was exclusively enriched within microglia, while epigenetic dysregulation predominantly fell within oligodendrocyte-specific H3K27ac regions. In addition, reanalysis of the original datasets using CHAS enabled identification of biological pathways associated with each neurological and psychiatric disorder at cellular resolution.
Collapse
Affiliation(s)
- Kitty B Murphy
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK.
| | - Yuqian Ye
- Department of Brain Sciences, Imperial College London, London, UK; UK Dementia Research Institute at Imperial College London, London, UK
| | - Maria Tsalenchuk
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, UK; UK Dementia Research Institute at Imperial College London, London, UK
| | - Sarah J Marzi
- UK Dementia Research Institute at King's College London, London, UK; Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Brain Sciences, Imperial College London, London, UK.
| |
Collapse
|
6
|
Ma S, Zhang T, Lv J, Liang S, Zhao S, Nan X, Dou Z, Yang J, Lu Y, Liu R, Li H. SORLA Orchestrates microglial dynamics for enhanced neuroprotection and recovery following ischemic stroke. Brain Behav Immun 2025:S0889-1591(25)00193-X. [PMID: 40389040 DOI: 10.1016/j.bbi.2025.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025] Open
Abstract
This study identifies a novel function of Sortilin-related receptor with A-type repeats (SORLA), traditionally linked to Alzheimer's Disease (AD) as a high-risk gene and associated with neuronal function, in modulating microglial responses to ischemic stroke. We discovered that SORLA expression is significantly reduced in microglia following stroke, a change linked to increased brain injury and diminished neurological recovery. Utilizing SORLA knockout and overexpression models, we demonstrated its essential role in adjusting microglial inflammatory responses. Notably, microglial-specific overexpression of SORLA not only promoted anti-inflammatory actions and effective phagocytosis but also surpassed traditional concepts of microglial polarization. This overexpression mitigated brain damage and enhanced neurofunctional recovery post-stroke, highlighting the neuroprotective potential of SORLA. This breakthrough challenges the prevailing understanding the role of SORLA and opens new therapeutic possibilities for stroke recovery, indicating its wider relevance for neurodegenerative disease management.
Collapse
Affiliation(s)
- Sehui Ma
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tongmei Zhang
- Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junkai Lv
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiqi Liang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuaizhu Zhao
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinyue Nan
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ziyue Dou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Yang
- Department of Rehabilitation, Wuhan Hankou Hospital, Wuhan 430010, China
| | - Youming Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hao Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Innovation Center for Brain Medical Sciences of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
7
|
Stanley N, Dhawka L, Jaikumar S, Huang YC, Zannas AS. Microglia Single-Cell RNA-Seq Enables Robust and Applicable Markers of Biological Aging. Aging Cell 2025:e70095. [PMID: 40371813 DOI: 10.1111/acel.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/27/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
"Biological aging clocks"-composite molecular markers thought to capture an individual's biological age-have been traditionally developed through bulk-level analyses of mixed cells and tissues. However, recent evidence highlights the importance of gaining single-cell-level insights into the aging process. Microglia are key immune cells in the brain shown to adapt functionally in aging and disease. Recent studies have generated single-cell RNA-sequencing (scRNA-seq) datasets that transcriptionally profile microglia during aging and development. Leveraging such datasets in humans and mice, we develop and compare computational approaches for generating transcriptome-wide summaries from microglia to establish robust and applicable aging clocks. Our results reveal that unsupervised, frequency-based summarization approaches, which encode distributions of cells across molecular subtypes, strike a balance in accuracy, interpretability, and computational efficiency. Notably, our computationally derived microglia markers achieve strong accuracy in predicting chronological age across three diverse single-cell datasets, suggesting that microglia exhibit characteristic changes in gene expression during aging and development that can be computationally summarized to create robust markers of biological aging. We further extrapolate and demonstrate the applicability of single-cell-based microglia clocks to readily available bulk RNA-seq data with an environmental input (early life stress), indicating the potential for broad utility of our models across genomic modalities and for testing hypotheses about how environmental inputs affect brain age. Such single-cell-derived markers can yield insights into the determinants of brain aging, ultimately promoting interventions that beneficially modulate health and disease trajectories.
Collapse
Affiliation(s)
- Natalie Stanley
- Department of Computer Science and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Luvna Dhawka
- Department of Computer Science and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Curriculum in Bioinformatics and Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sneha Jaikumar
- Department of Computer Science and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yu-Chen Huang
- Curriculum in Bioinformatics and Computational Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Anthony S Zannas
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Lau F, Binacchi R, Brugnara S, Cumplido-Mayoral A, Savino SD, Khan I, Orso A, Sartori S, Bellosta P, Carl M, Poggi L, Provenzano G. Using Single-Cell RNA sequencing with Drosophila, Zebrafish, and mouse models for studying Alzheimer's and Parkinson's disease. Neuroscience 2025; 573:505-517. [PMID: 40154937 DOI: 10.1016/j.neuroscience.2025.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Alzheimer's and Parkinson's disease are the most common neurodegenerative diseases, significantly affecting the elderly with no current cure available. With the rapidly aging global population, advancing research on these diseases becomes increasingly critical. Both disorders are often studied using model organisms, which enable researchers to investigate disease phenotypes and their underlying molecular mechanisms. In this review, we critically discuss the strengths and limitations of using Drosophila, zebrafish, and mice as models for Alzheimer's and Parkinson's research. A focus is the application of single-cell RNA sequencing, which has revolutionized the field by providing novel insights into the cellular and transcriptomic landscapes characterizing these diseases. We assess how combining animal disease modeling with high-throughput sequencing and computational approaches has advanced the field of Alzheimer's and Parkinson's disease research. Thereby, we highlight the importance of integrative multidisciplinary approaches to further our understanding of disease mechanisms and thus accelerating the development of successful therapeutic interventions.
Collapse
Affiliation(s)
- Frederik Lau
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Rebecca Binacchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Samuele Brugnara
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Alba Cumplido-Mayoral
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Serena Di Savino
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Ihsanullah Khan
- Department of Civil, Environmental and Mechanical Engineering, University of Trento 38123 Trento, Italy
| | - Angela Orso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Samuele Sartori
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy; Department of Medicine NYU Grossman School of Medicine, 550 First Avenue, 10016 NY, USA.
| | - Matthias Carl
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| | - Lucia Poggi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| |
Collapse
|
9
|
Augusto-Oliveira M, Arrifano GDP, Leal-Nazaré CG, Chaves-Filho A, Santos-Sacramento L, Lopes-Araujo A, Tremblay MÈ, Crespo-Lopez ME. Morphological diversity of microglia: Implications for learning, environmental adaptation, ageing, sex differences and neuropathology. Neurosci Biobehav Rev 2025; 172:106091. [PMID: 40049541 DOI: 10.1016/j.neubiorev.2025.106091] [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: 11/06/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Microglia are the brain resident macrophages that respond rapidly to any insult. These non-neuroectodermal cells are decorated with plenty of receptors allowing them to recognise and respond precisely to a multitude of stimuli. To do so, microglia undergo structural and functional changes aiming to actively keep the brain's homeostasis. However, some microglial responses, when sustained or exacerbated, can contribute to neuropathology and neurodegeneration. Many microglial molecular and cellular changes were identified that display a strong correlation with neuronal damage and neuroinflammation/disease status, as well as present key sex-related differences that modulate microglial outcomes. Nevertheless, the relationship between microglial structural and functional features is just beginning to be unravelled. Several reports show that microglia undergo soma and branch remodelling in response to environmental stimuli, ageing, neurodegenerative diseases, trauma, and systemic inflammation, suggesting a complex form and function link. Also, it is reasonable overall to suppose that microglia diminishing their process length and ramification also reduce their monitoring activity of synapses, which is critical for detecting any synaptic disturbance and performing synaptic remodelling. Elucidating the complex interactions between microglial morphological plasticity and its functional implications appears essential for the understanding of complex cognitive and behavioural processes in health and neuropathological conditions.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Amanda Lopes-Araujo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec, Qubec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada; College Member of the Royal Society of Canada, Canada.
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| |
Collapse
|
10
|
Wolf T, Moss L, Hudson C, Winters AM, Abdelmaboud SS, Avlas M, Wohlfahrt J, Guergues J, Bickford PC, Stevens SM. Chronic alcohol exposure during young adulthood attenuates microglial reactivity and downstream immune response pathways in a mouse model of tauopathy later in life. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:985-1000. [PMID: 40114609 PMCID: PMC12097940 DOI: 10.1111/acer.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by the buildup of amyloid-β and tau protein tangles. Alcohol use has been identified as a risk factor for AD; however, the molecular mechanisms underlying this potential causal link remain elusive. An emerging area of research focuses on the role of microglia, the brain's innate immune cells, in AD pathogenesis, with evidence suggesting that alcohol exposure may prime microglia to exhibit an exaggerated immune response when they are subsequently exposed to proinflammatory stimuli. METHODS We used a single 10-day chronic-plus-binge alcohol exposure model in male and female C57BL/6J mice aged 8-10 weeks One month later, tauopathy was induced via adenoviral vector (AAV)-mediated overexpression of h-p301L Tau. After 2.5 months, the mice underwent behavioral and cognitive testing. Two weeks later, microglia were collected using fluorescence-activated cell sorting (FACS) and processed for unbiased, mass spectrometry-based proteomic analysis to determine the molecular pathways related to microglial reactivity. RESULTS Microglia from mice exposed to alcohol in young adulthood exhibited a blunted immune response when challenged with AAV-mediated delivery and accumulation of human tau later in life. This was characterized by decreased expression of MHC II- and interferon-associated proteins and bioinformatic prediction of inhibited inflammation-related pathways in the absence of gross histological, behavioral, or cognitive deficits. These results demonstrate unique, temporally specific microglial reactivity to tau that is modulated by early adulthood alcohol exposure, implicating a microglial response that could negatively affect the mechanisms necessary for tau clearance and potentially exacerbate tau pathogenesis. CONCLUSIONS This study provides novel insights into the long-term effects of alcohol exposure in early adulthood on microglial function and the complexity of context-dependent microglial involvement in tauopathy. Consideration of early-adulthood environmental factors is critical for understanding and potentially mitigating the risk of neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Tiara Wolf
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Lauren Moss
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Charles Hudson
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Alexis M. Winters
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Salma S. Abdelmaboud
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Marta Avlas
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Jessica Wohlfahrt
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Jennifer Guergues
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Paula C. Bickford
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Stanley M. Stevens
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
11
|
Ma YN, Xia Y, Karako K, Song P, Tang W, Hu X. Decoding Alzheimer's Disease: Single-Cell Sequencing Uncovers Brain Cell Heterogeneity and Pathogenesis. Mol Neurobiol 2025:10.1007/s12035-025-04997-0. [PMID: 40304967 DOI: 10.1007/s12035-025-04997-0] [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: 10/30/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder marked by progressive cognitive decline and diverse neuropathological features. Recent advances in single-cell sequencing technologies have provided unprecedented insights into the cellular and molecular heterogeneity of the AD brain. This review systematically summarizes the applications of single-cell transcriptomic and epigenomic approaches in AD research, with a focus on the characterization of cell type- and subtype-specific transcriptomic alterations. This review highlights key discoveries related to selectively vulnerable neuronal and glial subpopulations, as well as transcriptional dysregulation associated with genetic risk loci such as APOE and TREM2. This review also discusses how the integration of single-cell RNA sequencing (scRNA-seq), assays for transposase-accessible chromatin using sequencing (ATAC-seq), and spatial transcriptomics elucidates disease trajectories and cellular communication networks across pathological stages. These insights not only enhance the understanding of the pathogenesis of AD but also pave the way for precision medicine through the identification of novel therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
- Integrated Neuroscience Center, Geriatric Hospital of Hainan, Haikou, 571100, China
| | - Kenji Karako
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Peipei Song
- Division of Global Health & Medicine, National Center for Global Health and Medicine, Tokyo, Japan.
- National College of Nursing, Tokyo, Japan.
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Global Health & Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
- Integrated Neuroscience Center, Geriatric Hospital of Hainan, Haikou, 571100, China.
| |
Collapse
|
12
|
Lopez-Atalaya JP, Bhojwani-Cabrera AM. Type I interferon signalling and interferon-responsive microglia in health and disease. FEBS J 2025. [PMID: 40299722 DOI: 10.1111/febs.70126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
Recent evidence suggests that type I interferon (IFN-I) signalling extends beyond its canonical roles in antiviral defence and immunomodulation. Over the past decade, dysregulated IFN-I signalling has been linked to genetic disorders and neurodegenerative diseases, where it may contribute to neurological impairments. Microglia have emerged as key mediators of IFN-I responses in the central nervous system. A distinct transcriptional state responsive to interferons has recently been identified in microglia. The activation of the IFN-I pathway in these cells is now recognised as pivotal in both development and neurodegeneration. This review is divided into two main sections: the first examines the broader role of IFN-I signalling in the central nervous system, particularly its contribution to neurological dysfunction; the second focuses on the specific state of interferon-responsive microglia, exploring its mechanisms and relevance in neurodegenerative conditions. Finally, we discuss how these areas intersect and their implications for both healthy and diseased states.
Collapse
Affiliation(s)
- Jose P Lopez-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| | - Aysha M Bhojwani-Cabrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
13
|
Sirimaharaj N, Thiankhaw K, Chattipakorn N, Chattipakorn SC. Unveiling the Protective Roles of Melatonin on Glial Cells in the Battle Against Alzheimer's Disease-Insights from In Vivo and In Vitro Studies. Mol Neurobiol 2025:10.1007/s12035-025-04904-7. [PMID: 40208552 DOI: 10.1007/s12035-025-04904-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a chronic, progressive neurodegenerative disorder that predominantly affects the elderly. Characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles, AD leads to memory loss, cognitive decline, and severe behavioral changes. As the most common form of dementia, AD imposes a significant global health burden, highlighting the need for interventions that address underlying disease mechanisms rather than only symptomatic treatment. Glial cells, including microglia and astrocytes, play a crucial role in AD progression by mediating neuroinflammatory responses and modulating Aβ clearance and neuronal health. Dysfunction in these cells can exacerbate neuroinflammation and neuronal damage, making glial cells an important target for therapeutic intervention. This review synthesizes findings from in vivo and in vitro studies on melatonin's effects on glial cell dysfunction in AD, emphasizing the multi-mechanistic nature of its neuroprotective properties. Recent studies highlight melatonin's potential as a therapeutic agent that addresses AD-related mechanisms through its interactions with glial cells. Melatonin has demonstrated protective effects, including reducing oxidative stress, apoptosis, and inflammation, inhibiting Aβ fibrillogenesis, and modulating amyloid precursor proteins. Additionally, its influence on glial cell activity, through melatonin receptor pathways, suggests it can alleviate neuroinflammation, a key component of AD progression. The collective evidence points to melatonin's promise as a therapeutic tool with potential roles in both preventive and adjunctive treatments for AD. However, further research is necessary to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Nopdanai Sirimaharaj
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kitti Thiankhaw
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
14
|
Zelic M, Blazier A, Pontarelli F, LaMorte M, Huang J, Tasdemir-Yilmaz OE, Ren Y, Ryan SK, Shapiro C, Morel C, Krishnaswami P, Levit M, Sood D, Chen Y, Gans J, Tang X, Hsiao-Nakamoto J, Huang F, Zhang B, Berry JD, Bangari DS, Gaglia G, Ofengeim D, Hammond TR. Single-cell transcriptomic and functional studies identify glial state changes and a role for inflammatory RIPK1 signaling in ALS pathogenesis. Immunity 2025; 58:961-979.e8. [PMID: 40132594 DOI: 10.1016/j.immuni.2025.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron loss. Microglia and astrocyte-driven neuroinflammation is prominent in ALS, but the cell state dynamics and pathways driving disease remain unclear. We performed single-nucleus RNA sequencing of ALS spinal cords and identified altered glial cell states, including increased expression of inflammatory and glial activation markers. Many of these signals converged on the inflammation and cell death regulator receptor-interacting protein kinase 1 (RIPK1) and the necroptotic cell death pathway. In superoxide dismutase 1 (SOD1)G93A mice, blocking RIPK1 kinase activity delayed symptom onset and motor impairment and modulated glial responses. We used human induced pluripotent stem cell (iPSC)-derived motor neuron, astrocyte, and microglia tri-cultures to identify potential biomarkers that are secreted upon RIPK1 activation in vitro and modulated by RIPK1 inhibition in the cerebrospinal fluid (CSF) of people with ALS. These data reveal ALS-enriched glial populations associated with inflammation and suggest a deleterious role for neuroinflammatory signaling in ALS pathogenesis.
Collapse
Affiliation(s)
- Matija Zelic
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA.
| | - Anna Blazier
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | | | - Michael LaMorte
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Jeremy Huang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | - Yi Ren
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Sean K Ryan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Cynthia Shapiro
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Caroline Morel
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | | | - Mikhail Levit
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Disha Sood
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA 02141, USA
| | - Yao Chen
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Joseph Gans
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - Xinyan Tang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | | | - Fen Huang
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Bailin Zhang
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | - James D Berry
- Healey Center for ALS, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dinesh S Bangari
- Sanofi, Global Discovery Pathology and Multimodal Imaging, Cambridge, MA 02141, USA
| | - Giorgio Gaglia
- Sanofi, Precision Medicine and Computational Biology, Cambridge, MA 02141, USA
| | | | | |
Collapse
|
15
|
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
|
16
|
Reitsema VA, Schreuder L, Gerrits E, Eggen BJL, Goris M, Laman JD, de Rooij SE, Wesseling EM, Bouma HR, Henning RH. Calorie restriction increases the sensitivity of progeroid Ercc1 Δ/- mice to acute (neuro)inflammation. GeroScience 2025; 47:1641-1652. [PMID: 39287878 PMCID: PMC11978592 DOI: 10.1007/s11357-024-01347-1] [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: 06/07/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Hospitalized elderly patients frequently suffer from delirium, especially in the context of sepsis-associated encephalopathy. Current treatments of delirium are merely symptomatic. Calorie restriction (CR) is both a promising strategy to protect against sepsis and has beneficial effects on aging-induced neurodegeneration. In this study, we investigated whether six weeks of 30% CR had protective effects on lipopolysaccharide (LPS) induced (neuro)inflammation in wild-type (WT) and progeroid mice deficient in the DNA excision-repair gene Ercc1 (Ercc1Δ/-). While CR did not affect the LPS-induced inflammatory response in WT mice, CR exaggerated the peripheral inflammatory response in Ercc1Δ/- mice, as evidenced by an increase of pro-inflammatory serum cytokines (TNF-α, IL-1β, and IFN-γ) and kidney injury marker Ngal. Neuroinflammatory effects were assessed by RNA-sequencing of isolated microglia. Similarly, CR did not affect microglia gene expression in WT mice, but increased neuroinflammation-associated gene expression in Ercc1Δ/- mice. In conclusion, CR increases the peripheral and brain inflammatory response of Ercc1Δ/- mice to a systemic inflammatory stimulus.
Collapse
Affiliation(s)
- V A Reitsema
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - L Schreuder
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E Gerrits
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Goris
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J D Laman
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S E de Rooij
- Department of Internal Medicine, University Center for Geriatric Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E M Wesseling
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - H R Bouma
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- Department of Acute Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - R H Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
17
|
Hu W, Chen M, Lin Y, Zhang H, Sun L, Shao W, Ye Y, Cheng Y, Zhou S, Hu P, Wu X, Xu Y, Wang K. Neuronal CD47 induces behavioral alterations and ameliorates microglial synaptic pruning in wild-type and Alzheimer's mouse models. Cell Biosci 2025; 15:38. [PMID: 40140948 PMCID: PMC11948738 DOI: 10.1186/s13578-025-01378-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Microglia are brain-resident macrophages that play a crucial role in synapse pruning during the development and progression of various neuropsychiatric disorders, including autism spectrum disorder (ASD) and Alzheimer's disease (AD). Mechanistically, CD47 protein acts as a potent 'do not eat me' signal, protecting synapses from phagocytosis by microglia. However, the functional role of the upregulated neuronal CD47 signal under both physiological and pathological conditions remains unclear. RESULTS We utilized an adeno-associated virus gene expression system to induce neuron-specific overexpression of CD47 in wild-type and 5xFAD mice, assessing its effects on microglial synaptic phagocytosis and mouse behaviors. Our results indicate that neuronal CD47 induces ASD-like behaviors and synaptic pruning defects, while promoting behavioral disinhibition and improving memory in wild-type mice. Single-nucleus RNA sequencing was employed to profile gene expression patterns in subpopulations of neurons and microglia. Notably, neuronal CD47 enhances synaptic pathways in neurons and particularly shifts microglial subpopulations from a disease-associated state to a homeostatic state. Additionally, neuronal CD47 reduces excessive microglial synaptic phagocytosis induced by Aβ pathology in 5xFAD mice. CONCLUSION Our study provides evidence that neuronal CD47 overexpression results in synaptic pruning defects and is involved in the pathogenesis of ASD, while also playing a beneficial role in mitigating excessive synaptic loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Mengting Chen
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuxue Lin
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Li Sun
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Wei Shao
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yuping Ye
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Yujie Cheng
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China
| | - Yin Xu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230022, China.
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, 230032, China.
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, 230032, China.
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, 230032, China.
- Laboratory of Molecular Neuropsychiatry, Anhui Medical University, Hefei, 230032, China.
- Anhui Institute of Translational Medicine, Hefei, 230032, China.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Qiu M, Zhao L, Li X, Fan Y, Liu M, Hua D, Zhu Y, Liang Y, Zhang Y, Xiao W, Xu X, Li J. Decoding dengue's neurological assault: insights from single-cell CNS analysis in an immunocompromised mouse model. J Neuroinflammation 2025; 22:62. [PMID: 40038739 PMCID: PMC11877810 DOI: 10.1186/s12974-025-03383-w] [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: 12/10/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Dengue encephalitis, a severe neurological complication of dengue virus infection, is increasingly recognized for its rising incidence and significant public health burden. Despite its growing prevalence, the underlying mechanisms and effective therapeutic strategies remain poorly understood. METHODS Cellular atlas of dengue encephalitis was determined by single-nucleus RNA sequencing. Viral load of dengue virus and the level of cytokines expression was detected by RT-qPCR. The target cells of dengue virus were verified by immunofluorescence. The cytotoxic effect of CD8+ T cell was determined by flow cytometry, immunofluorescence, in vivo CD8+ T cell depletion, adoptive transfer and CCK-8-based cell viability assay. Axonal and synaptic reduction induced by dengue virus infection was demonstrated by RT-qPCR, Western blot, transmission electron microscope and immunofluorescence. Finally, motor and sensory functions of mice were detected by open field test and hot plate test, respectively. RESULTS In this study, we utilized single-nucleus RNA sequencing on brain tissues from a dengue-infected murine model to construct a comprehensive cellular atlas of dengue encephalitis. Our findings identify neurons, particularly inhibitory GABAergic subtypes, as the primary targets of dengue virus. Additionally, immune cell infiltration was observed, contributing to significant neurological damage. Comprehensive analyses of cell-cell communication, combined with CD8+ T cell depletion and transfer restoration experiments, have elucidated the critical role of CD8+ T cells in triggering encephalitis through their interaction with neurons. These cells infiltrate the brain from peripheral circulation, interact with neurons, and induce damage of synapse and axon, accompanied by neurological dysfunction. CONCLUSION We defined cellular atlas of dengue encephalitis in mouse model and identified the primary target neuron of dengue virus. In addition, we demonstrated the significant cytotoxic effect of CD8+ T cell, which leads to apoptosis of neuron and neurological dysfunction of mice. Our study provides a molecular and cellular framework for understanding dengue encephalitis through advanced sequencing technologies. The insights gained serve as a foundation for future investigations into its pathogenesis and the development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Minyue Qiu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Lixin Zhao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Xiaojia Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yipei Fan
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Minchi Liu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Dong Hua
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yunkai Zhu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yinyin Liang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Wen Xiao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiaofeng Xu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Jintao Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China.
- Institute of Immunology, Army Medical University, Chongqing, China.
| |
Collapse
|
21
|
Zhang G, Yao Q, Long C, Yi P, Song J, Wu L, Wan W, Rao X, Lin Y, Wei G, Ying J, Hua F. Infiltration by monocytes of the central nervous system and its role in multiple sclerosis: reflections on therapeutic strategies. Neural Regen Res 2025; 20:779-793. [PMID: 38886942 PMCID: PMC11433895 DOI: 10.4103/nrr.nrr-d-23-01508] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 02/18/2024] [Indexed: 06/20/2024] Open
Abstract
Mononuclear macrophage infiltration in the central nervous system is a prominent feature of neuroinflammation. Recent studies on the pathogenesis and progression of multiple sclerosis have highlighted the multiple roles of mononuclear macrophages in the neuroinflammatory process. Monocytes play a significant role in neuroinflammation, and managing neuroinflammation by manipulating peripheral monocytes stands out as an effective strategy for the treatment of multiple sclerosis, leading to improved patient outcomes. This review outlines the steps involved in the entry of myeloid monocytes into the central nervous system that are targets for effective intervention: the activation of bone marrow hematopoiesis, migration of monocytes in the blood, and penetration of the blood-brain barrier by monocytes. Finally, we summarize the different monocyte subpopulations and their effects on the central nervous system based on phenotypic differences. As activated microglia resemble monocyte-derived macrophages, it is important to accurately identify the role of monocyte-derived macrophages in disease. Depending on the roles played by monocyte-derived macrophages at different stages of the disease, several of these processes can be interrupted to limit neuroinflammation and improve patient prognosis. Here, we discuss possible strategies to target monocytes in neurological diseases, focusing on three key aspects of monocyte infiltration into the central nervous system, to provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangyong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qing Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Chubing Long
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
22
|
Prater KE, Lin KZ. All the single cells: Single-cell transcriptomics/epigenomics experimental design and analysis considerations for glial biologists. Glia 2025; 73:451-473. [PMID: 39558887 PMCID: PMC11809281 DOI: 10.1002/glia.24633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024]
Abstract
Single-cell transcriptomics, epigenomics, and other 'omics applied at single-cell resolution can significantly advance hypotheses and understanding of glial biology. Omics technologies are revealing a large and growing number of new glial cell subtypes, defined by their gene expression profile. These subtypes have significant implications for understanding glial cell function, cell-cell communications, and glia-specific changes between homeostasis and conditions such as neurological disease. For many, the training in how to analyze, interpret, and understand these large datasets has been through reading and understanding literature from other fields like biostatistics. Here, we provide a primer for glial biologists on experimental design and analysis of single-cell RNA-seq datasets. Our goal is to further the understanding of why decisions are made about datasets and to enhance biologists' ability to interpret and critique their work and the work of others. We review the steps involved in single-cell analysis with a focus on decision points and particular notes for glia. The goal of this primer is to ensure that single-cell 'omics experiments continue to advance glial biology in a rigorous and replicable way.
Collapse
Affiliation(s)
- Katherine E. Prater
- Department of Neurology, University of Washington School of Medicine, Seattle 98195
| | - Kevin Z. Lin
- Department of Biostatistics, University of Washington, Seattle 98195
| |
Collapse
|
23
|
Abellanas MA, Purnapatre M, Burgaletto C, Schwartz M. Monocyte-derived macrophages act as reinforcements when microglia fall short in Alzheimer's disease. Nat Neurosci 2025; 28:436-445. [PMID: 39762659 DOI: 10.1038/s41593-024-01847-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/06/2024] [Indexed: 03/12/2025]
Abstract
The central nervous system (CNS) is endowed with its own resident innate immune cells, the microglia. They constitute approximately 10% of the total cells within the CNS parenchyma and act as 'sentinels', sensing and mitigating any deviation from homeostasis. Nevertheless, under severe acute or chronic neurological injury or disease, microglia are unable to contain the damage, and the reparative activity of monocyte-derived macrophages (MDMs) is required. The failure of the microglia under such conditions could be an outcome of their prolonged exposure to hostile stimuli, leading to their exhaustion or senescence. Here, we describe the conditions under which the microglia fall short, focusing mainly on the context of Alzheimer's disease, and shed light on the functions performed by MDMs. We discuss whether and how MDMs engage in cross-talk with the microglia, why their recruitment is often inadequate, and potential ways to augment their homing to the brain in a well-controlled manner.
Collapse
Affiliation(s)
- Miguel A Abellanas
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Chiara Burgaletto
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
24
|
Wang H, Fleishman JS, Wu S, Wang G, Du L, Li J, Du J. cGAS-STING targeting offers novel therapeutic opportunities in neurological diseases. Ageing Res Rev 2025; 105:102691. [PMID: 39954791 DOI: 10.1016/j.arr.2025.102691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/10/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS) is a cytosolic DNA sensor that produces the secondary messenger cGAMP. cGAMP activates the endoplasmic reticulum-associated adaptor stimulator of interferon genes (STING) and activates the innate immune system to produce a type I interferon response. Besides sensing microbial DNA, cGAS can also be activated by self-DNA or endogenous DNA, including that derived from genotoxic extranuclear chromatin and mitochondrially released DNA, indicating that cGAS-STING is an important mechanism in sterile inflammatory responses, autoimmunity, and cellular senescence. However, aberrant activation of the cGAS-STING pathway results in inflammatory and autoimmune diseases. cGAS-STING has emerged as a vital mechanism driving the pathogenesis of inflammation, implicating cGAS-STING signaling in neurological diseases. In this review, we first outline the principal elements of the cGAS-STING signaling cascade, summarizing recent research highlighting how cGAS-STING activation contributes to the pathogenesis of neurological diseases, including various autoimmune, autoinflammatory, and neurodegenerative diseases. Next, we outline selective small-molecule modulators that function as cGAS-STING inhibitors and summarize their mechanisms for treating multiple neurological diseases. Finally, we discuss key limitations of the current therapeutic paradigm and generate possible strategies to overcome them.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Guan Wang
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Lida Du
- Division of Neurobiology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Jilai Li
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China.
| | - Jichen Du
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
25
|
Reid AN, Jayadev S, Prater KE. Microglial Responses to Alzheimer's Disease Pathology: Insights From "Omics" Studies. Glia 2025; 73:519-538. [PMID: 39760224 PMCID: PMC11801359 DOI: 10.1002/glia.24666] [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: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
Human genetics studies lent firm evidence that microglia are key to Alzheimer's disease (AD) pathogenesis over a decade ago following the identification of AD-associated genes that are expressed in a microglia-specific manner. However, while alterations in microglial morphology and gene expression are observed in human postmortem brain tissue, the mechanisms by which microglia drive and contribute to AD pathology remain ill-defined. Numerous mouse models have been developed to facilitate the disambiguation of the biological mechanisms underlying AD, incorporating amyloidosis, phosphorylated tau, or both. Over time, the use of multiple technologies including bulk tissue and single cell transcriptomics, epigenomics, spatial transcriptomics, proteomics, lipidomics, and metabolomics have shed light on the heterogeneity of microglial phenotypes and molecular patterns altered in AD mouse models. Each of these 'omics technologies provide unique information and biological insight. Here, we review the literature on the approaches and findings of these methods and provide a synthesis of the knowledge generated by applying these technologies to mouse models of AD.
Collapse
Affiliation(s)
- Aquene N. Reid
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195
| | - Suman Jayadev
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98195
| | - Katherine E. Prater
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
26
|
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
|
27
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Thompson JR, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. Cell Rep 2025; 44:115300. [PMID: 40009515 DOI: 10.1016/j.celrep.2025.115300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/19/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we present transcriptome-wide spatial gene expression maps of the human dentate gyrus and investigate age-associated changes across the lifespan. Genes associated with neurogenesis and the extracellular matrix are enriched in infants and decline throughout development and maturation. Following infancy, inhibitory neuron markers increase, and cellular proliferation markers decrease. We also identify spatio-molecular signatures that support existing evidence for protracted maturation of granule cells during adulthood and age-associated increases in neuroinflammation-related gene expression. Our findings support the notion that the hippocampal neurogenic niche undergoes major changes following infancy and identify molecular regulators of brain aging in glial- and neuropil-enriched tissue.
Collapse
Affiliation(s)
- Anthony D Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Alexis R Papariello
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Ryan A Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Erik D Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jacqueline R Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD 21205, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Center for Computational Biology, Johns Hopkins University, Baltimore, MD 21205, USA; Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
28
|
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
|
29
|
Balkhi S, Di Spirito A, Poggi A, Mortara L. Immune Modulation in Alzheimer's Disease: From Pathogenesis to Immunotherapy. Cells 2025; 14:264. [PMID: 39996737 PMCID: PMC11853524 DOI: 10.3390/cells14040264] [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: 12/25/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia, affecting a significant proportion of the elderly population. AD is characterized by cognitive decline and functional impairments due to pathological hallmarks like amyloid β-peptide (Aβ) plaques and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau. Microglial activation, chronic neuroinflammation, and disruptions in neuronal communication further exacerbate the disease. Emerging research suggests that immune modulation could play a key role in AD treatment given the significant involvement of neuroinflammatory processes. This review focuses on recent advancements in immunotherapy strategies aimed at modulating immune responses in AD, with a specific emphasis on microglial behavior, amyloid clearance, and tau pathology. By exploring these immunotherapeutic approaches, we aim to provide insights into their potential to alter disease progression and improve patient outcomes, contributing to the evolving landscape of AD treatment.
Collapse
Affiliation(s)
- Sahar Balkhi
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (A.D.S.); (L.M.)
| | - Anna Di Spirito
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (A.D.S.); (L.M.)
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (A.D.S.); (L.M.)
| |
Collapse
|
30
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Le LHD, Eliseeva S, Plunk E, Kara-Pabani K, Li H, Yarovinsky F, Majewska AK. The microglial response to inhibition of Colony-stimulating-factor-1 receptor by PLX3397 differs by sex in adult mice. Cell Rep 2025; 44:115176. [PMID: 39842435 PMCID: PMC11877653 DOI: 10.1016/j.celrep.2024.115176] [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: 04/08/2024] [Revised: 10/08/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Microglia, the resident macrophages of the brain, are derived from the yolk sac and colonize the brain before the blood-brain barrier forms. Once established, they expand locally and require Colony-stimulating-factor-1 receptor (CSF1R) signaling for their development and maintenance. CSF1R inhibitors have been used extensively to deplete microglia in the healthy and diseased brain. In this study, we demonstrated sex-dependent differences in the microglial response to the CSF1R inhibitor PLX3397. Male mice exhibited greater microglial depletion compared to females. Transcriptomic and flow cytometry analysis revealed sex-specific differences in the remaining microglia population, with female microglia upregulating autophagy and proteostasis pathways while male microglia increased mitobiogenesis. Furthermore, manipulating key microglial receptors by using different transgenic mouse lines resulted in changes in depletion efficacies that were also sex dependent. These findings suggest sex-dependent microglial survival mechanisms, which might contribute to the well-documented sex differences in various neurological disorders.
Collapse
Affiliation(s)
- Linh H D Le
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Sophia Eliseeva
- Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Elizabeth Plunk
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA; Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Kallam Kara-Pabani
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA
| | - Herman Li
- Medical Scientist Training Program, University of Rochester, Rochester, NY 14642, USA
| | - Felix Yarovinsky
- Department of Microbiology and Immunology, Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester, Rochester, NY 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
32
|
Rao A, Chen N, Kim MJ, Blumenfeld J, Yip O, Liang Z, Shostak D, Hao Y, Nelson MR, Koutsodendris N, Grone B, Ding L, Yoon SY, Arriola P, Zilberter M, Huang Y. Microglia depletion reduces human neuronal APOE4-related pathologies in a chimeric Alzheimer's disease model. Cell Stem Cell 2025; 32:86-104.e7. [PMID: 39500314 DOI: 10.1016/j.stem.2024.10.005] [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: 11/16/2023] [Revised: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 11/13/2024]
Abstract
Despite strong evidence supporting the important roles of both apolipoprotein E4 (APOE4) and microglia in Alzheimer's disease (AD) pathogenesis, the effects of microglia on neuronal APOE4-related AD pathogenesis remain elusive. To examine such effects, we utilized microglial depletion in a chimeric model with induced pluripotent stem cell (iPSC)-derived human neurons in mouse hippocampus. Specifically, we transplanted homozygous APOE4, isogenic APOE3, and APOE-knockout (APOE-KO) iPSC-derived human neurons into the hippocampus of human APOE3 or APOE4 knockin mice and then depleted microglia in half of the chimeric mice. We found that both neuronal APOE and microglial presence were important for the formation of Aβ and tau pathologies in an APOE isoform-dependent manner (APOE4 > APOE3). Single-cell RNA sequencing analysis identified two pro-inflammatory microglial subtypes with elevated MHC-II gene expression enriched in chimeric mice with human APOE4 neuron transplants. These findings highlight the concerted roles of neuronal APOE, especially APOE4, and microglia in AD pathogenesis.
Collapse
Affiliation(s)
- Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nuo Chen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Zherui Liang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - David Shostak
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Leo Ding
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
33
|
Henry RJ, Loane DJ. Unraveling the complexity of microglial responses in traumatic brain and spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:113-132. [PMID: 40148040 DOI: 10.1016/b978-0-443-19102-2.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Microglia, the resident innate immune cells of the central nervous system (CNS), play an important role in neuroimmune signaling, neuroprotection, and neuroinflammation. In the healthy CNS, microglia adopt a surveillant and antiinflammatory phenotype characterized by a ramified scanning morphology that maintains CNS homeostasis. In response to acquired insults, such as traumatic brain injury (TBI) or spinal cord injury (SCI), microglia undergo a dramatic morphologic and functional switch to that of a reactive state. This microglial switch is initially protective and supports the return of the injured tissue to a physiologic homeostatic state. However, there is now a significant body of evidence that both TBI and SCI can result in a chronic state of microglial activation, which contributes to neurodegeneration and impairments in long-term neurologic outcomes in humans and animal models. In this review, we discuss the complex role of microglia in the pathophysiology of TBI and SCI, and recent advancements in knowledge of microglial phenotypic states in the injured CNS. Furthermore, we highlight novel therapeutic strategies targeting chronic microglial responses in experimental models and discuss how they may ultimately be translated to the clinic for human brain and SCI.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Pharmacology, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Huang X, Jannu AJ, Song Z, Jury‐Garfe N, Lasagna‐Reeves CA, Alzheimer's Disease Neuroimaging Initiative, Johnson TS, Huang K, Zhang J. Predicting Alzheimer's disease subtypes and understanding their molecular characteristics in living patients with transcriptomic trajectory profiling. Alzheimers Dement 2025; 21:e14241. [PMID: 39812331 PMCID: PMC11772740 DOI: 10.1002/alz.14241] [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: 05/06/2024] [Revised: 07/18/2024] [Accepted: 08/02/2024] [Indexed: 01/16/2025]
Abstract
INTRODUCTION Deciphering the diverse molecular mechanisms in living Alzheimer's disease (AD) patients is a big challenge but is pivotal for disease prognosis and precision medicine development. METHODS Utilizing an optimal transport approach, we conducted graph-based mapping of transcriptomic profiles to transfer AD subtype labels from ROSMAP monocyte samples to ADNI and ANMerge peripheral blood mononuclear cells. Subsequently, differential expression followed by comparative pathway and diffusion pseudotime analysis were applied to each cohort to infer the progression trajectories. Survival analysis with real follow-up time was used to obtain potential biomarkers for AD prognosis. RESULTS AD subtype labels were accurately transferred onto the blood samples of ADNI and ANMerge living patients. Pathways and associated genes in neutrophil degranulation-like immune process, immune acute phase response, and IL-6 signaling were significantly associated with AD progression. DISCUSSION The work enhanced our understanding of AD progression in different subtypes, offering insights into potential biomarkers and personalized interventions for improved patient care. HIGHLIGHTS We applied an innovative optimal transport-based approach to map transcriptomic data from different Alzheimer's disease (AD) cohort studies and transfer known AD subtype labels from ROSMAP monocyte samples to peripheral blood mononuclear cell (PBMC) samples within ADNI and ANMerge cohorts. Through comprehensive trajectory and comparative analysis, we investigated the molecular mechanisms underlying different disease progression trajectories in AD. We validated the accuracy of our AD subtype label transfer and identified prognostic genetic markers associated with disease progression, facilitating personalized treatment strategies. By identifying and predicting distinctive AD subtypes and their associated pathways, our study contributes to a deeper understanding of AD heterogeneity.
Collapse
Affiliation(s)
- Xiaoqing Huang
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Asha Jacob Jannu
- Department of Biohealth InformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ziyan Song
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Nur Jury‐Garfe
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Cristian A. Lasagna‐Reeves
- Department of Anatomy, Cell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Travis S. Johnson
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kun Huang
- Department of Biostatistics & Health Data ScienceIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jie Zhang
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
35
|
Bortolin V, Mansuroglu Z, Conquet L, Calcagno G, Lambert F, Marin-Obando JP, Segrt H, Savino M, Menidjel R, Souès S, Buée L, Niedergang F, Galas MC, Montagutelli X, Bonnefoy E. Protein kinase R induced by type I interferons is a main regulator of reactive microglia in Zika virus infection. Glia 2025; 73:80-104. [PMID: 39359232 DOI: 10.1002/glia.24619] [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: 04/26/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Microglial cells are the phagocytic cells of the brain that under physiological conditions participate in brain homeostasis and surveillance. Under pathogenic states, microglia undergoes strong morphological and transcriptional changes potentially leading to sustained neuroinflammation, brain damage, and cognitive disorders. Postnatal and adult Zika virus (ZIKV) brain infection is characterized by the induction of reactive microglia associated with brain inflammation, synapse loss and neuropathogenesis. Contrary to neurons, microglial cells are not infected by ZIKV thus raising the question of the mechanism governing ZIKV-induced microglia's reactivity. In this work, we have questioned the role of exogenous, neuronal type I interferons (IFNs-I) in regulating ZIKV-induced microglia's reactivity. Primary cultured microglial cells were either treated with conditioned media from ZIKV-infected mature neurons or co-cultured with ZIKV-infected neurons. Using either an antibody directed against the IFNAR receptor that neutralizes the IFNs-I response or Ifnar-/-microglial cells, we demonstrate that IFNs-I produced by ZIKV-infected neurons are the main regulators of the phagocytic capacity and the pro-inflammatory gene expression profile of reactive, non-infected microglial cells. We identify protein kinase R (PKR), whose expression is activated by IFNs-I, as a major regulator of the phagocytic capacity, pro-inflammatory response, and morphological changes of microglia induced by IFNs-I while up-regulating STAT1 phosphorylation and IRF1 expression. Results obtained herein in vitro with primary cultured cells and in vivo in ZIKV-infected adult immunocompetent mice, unravel a role for IFNs-I and PKR in directly regulating microglia's reactivity that could be at work in other infectious and non-infectious brain pathologies.
Collapse
Affiliation(s)
| | - Zeyni Mansuroglu
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Laurine Conquet
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Gaetano Calcagno
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Fanny Lambert
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | | | - Helena Segrt
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Mary Savino
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Reyene Menidjel
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Sylvie Souès
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Luc Buée
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | | | - Marie-Christine Galas
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Eliette Bonnefoy
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| |
Collapse
|
36
|
Medd M. TREM2 in Regulating Macrophage Inflammatory Responses and Disease Pathogenesis. Crit Rev Immunol 2025; 45:15-24. [PMID: 39976515 DOI: 10.1615/critrevimmunol.2024054889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is a cell surface receptor belonging to the TREM family that is predominantly expressed on myeloid cells such as granulocytes, monocytes, osteocytes, macrophages, and microglia. Although much of the functionality of TREM2 is not well understood at the molecular level, it is well established that TREM2 plays a significant role in the regulation of a broad definition of macrophage inflammatory responses. Dysregulation of TREM2 has been implicated in a large number of diseases including Alzheimer's disease, Nasu-Hakola disease, bone-related diseases, and atherosclerosis. The TREM2 gene is highly conserved evolutionarily and at the level of controlling its expression. The function of TREM2 is highly conserved across the broad definition of macrophages, including microglia, osteoclasts, and vascular macrophages. This genetic and physiological "niche conservatism" strongly suggests its pivotal role in regulating inflammatory responses. This mini-review summarizes our current understanding of the structure, expression, and function of TREM2 in the pathogenesis of macrophage-mediated diseases.
Collapse
|
37
|
Anton PE, Maphis NM, Linsenbardt DN, Coleman LG. Excessive Alcohol Use as a Risk Factor for Alzheimer's Disease: Epidemiological and Preclinical Evidence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:211-242. [PMID: 40128481 DOI: 10.1007/978-3-031-81908-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Alcohol use has recently emerged as a modifiable risk factor for Alzheimer's disease (AD). However, the neurobiological mechanisms by which alcohol interacts with AD pathogenesis remain poorly understood. In this chapter, we review the epidemiological and preclinical support for the interaction between alcohol use and AD. We hypothesize that alcohol use increases the rate of accumulation of specific AD-relevant pathologies during the prodromal phase and exacerbates dementia onset and progression. We find that alcohol consumption rates are increasing in adolescence, middle age, and aging populations. In tandem, rates of AD are also on the rise, potentially as a result of this increased alcohol use throughout the lifespan. We then review the biological processes in common between alcohol use disorder and AD as a means to uncover potential mechanisms by which they interact; these include oxidative stress, neuroimmune function, metabolism, pathogenic tauopathy development and spread, and neuronal excitatory/inhibitory balance (EIB). Finally, we provide some forward-thinking suggestions we believe this field should consider. In particular, the inclusion of alcohol use assessments in longitudinal studies of AD and more preclinical studies on alcohol's impacts using better animal models of late-onset Alzheimer's disease (LOAD).
Collapse
Affiliation(s)
- Paige E Anton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Nicole M Maphis
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - David N Linsenbardt
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
38
|
Demmings MD, da Silva Chagas L, Traetta ME, Rodrigues RS, Acutain MF, Barykin E, Datusalia AK, German-Castelan L, Mattera VS, Mazengenya P, Skoug C, Umemori H. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J Neurochem 2025; 169:e16258. [PMID: 39680483 DOI: 10.1111/jnc.16258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Neuron-glia interactions are fundamental to the development and function of the nervous system. During development, glia, including astrocytes, microglia, and oligodendrocytes, influence neuronal differentiation and migration, synapse formation and refinement, and myelination. In the mature brain, glia are crucial for maintaining neural homeostasis, modulating synaptic activity, and supporting metabolic functions. Neurons, inherently vulnerable to various stressors, rely on glia for protection and repair. However, glia, in their reactive state, can also promote neuronal damage, which contributes to neurodegenerative and neuropsychiatric diseases. Understanding the dual role of glia-as both protectors and potential aggressors-sheds light on their complex contributions to disease etiology and pathology. By appropriately modulating glial activity, it may be possible to mitigate neurodegeneration and restore neuronal function. In this review, which originated from the International Society for Neurochemistry (ISN) Advanced School in 2019 held in Montreal, Canada, we first describe the critical importance of glia in the development and maintenance of a healthy nervous system as well as their contributions to neuronal damage and neurological disorders. We then discuss potential strategies to modulate glial activity during disease to protect and promote a properly functioning nervous system. We propose that targeting glial cells presents a promising therapeutic avenue for rebuilding the nervous system.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Luana da Silva Chagas
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marianela E Traetta
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Rui S Rodrigues
- University of Bordeaux, INSERM, Neurocentre Magendie U1215, Bordeaux, France
| | - Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Evgeny Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER Raebareli), Raebareli, UP, India
| | - Liliana German-Castelan
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vanesa S Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB-FFyB-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedzisai Mazengenya
- Center of Medical and bio-Allied Health Sciences Research, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Cecilia Skoug
- Department of Neuroscience, Physiology & Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, UK
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Dufour A, Heydari Olya A, Foulon S, Réda C, Mokhtari A, Faivre V, Hua J, Bokobza C, Griffiths AD, Nghe P, Gressens P, Delahaye-Duriez A, Van Steenwinckel J. Neonatal inflammation impairs developmentally-associated microglia and promotes a highly reactive microglial subset. Brain Behav Immun 2025; 123:466-482. [PMID: 39322088 DOI: 10.1016/j.bbi.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
Microglia and border-associated macrophages play critical roles in both immunity and neurodevelopment. The disruption of microglial development trajectories by neonatal inflammation is an important issue in research on neurodevelopmental disorders (NDDs), as models have suggested a strong association between inflammation and cognitive deficits. Here, we explored by single-cell RNA sequencing and flow cytometry the impact of neonatal inflammation in a mouse NDD model on brain myeloid cell subsets. A specific subset of microglia expressing the complement receptor C5ar1 has been identified, in which inflammatory pathways are most strongly activated. Based on transcriptional similarity, this subset appears to originate from the most mature and "homeostatic" microglia at this stage of development and demonstrated hypersensitivity to inflammation. Besides that, Spp1-microglia supporting oligodendrocyte differentiation, primitive and proliferative microglia were reduced by inflammation. These findings suggest major changes in microglial subsets developmental trajectories and reactivity contributing to NDDs induced by neonatal inflammation.
Collapse
Affiliation(s)
- Adrien Dufour
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France; Université Paris Saclay, INRAE, AgroParisTech, GABI, Domaine de Vilvert, 78350 Jouy en Josas, France
| | | | - Sophie Foulon
- Laboratoire de Biochimie, UMR CBI 8231, ESPCI Paris,10 rue Vauquelin 75005 Paris, France
| | - Clémence Réda
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France
| | | | - Valérie Faivre
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France
| | - Jennifer Hua
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France
| | - Cindy Bokobza
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France
| | - Andrew D Griffiths
- Laboratoire de Biochimie, UMR CBI 8231, ESPCI Paris,10 rue Vauquelin 75005 Paris, France
| | - Philippe Nghe
- Laboratoire de Biochimie, UMR CBI 8231, ESPCI Paris,10 rue Vauquelin 75005 Paris, France; Laboratoire Biophysique et Evolution, UMR CBI 8231, ESPCI Paris,10 rue Vauquelin 75005 Paris, France
| | | | - Andrée Delahaye-Duriez
- NeuroDiderot, INSERM, Université Paris Cité, Paris, France; Unité fonctionnelle de médecine génomique et génétique clinique, Hôpital Jean Verdier, AP-HP, 93140 Bondy, France; Université Sorbonne Paris Nord, UFR de santé, médecine et biologie humaine, 93000 Bobigny, France.
| | | |
Collapse
|
40
|
Chiu Y, Xia S, Qiao H, Zhao Z. Genetically Engineered Mouse Models for Alzheimer Disease and Frontotemporal Dementia: New Insights from Single-Cell and Spatial Transcriptomics. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00447-4. [PMID: 39743215 DOI: 10.1016/j.ajpath.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
Abstract
Neurodegenerative diseases, including Alzheimer disease, frontotemporal dementia, Parkinson disease, Huntington disease, and amyotrophic lateral sclerosis, are often casually linked to protein aggregation and inclusion. As the origins of those proteinopathies have been biochemically traced and genetically mapped, genetically engineered animal models carrying the specific mutations or variants are widely used for investigating the etiology of these diseases, as well as for testing potential therapeutics. This article focuses on the mouse models of Alzheimer disease and closely related frontotemporal dementia, particularly the ones that have provided most valuable knowledge, or are in a trajectory of doing so. More importantly, some of the major findings from these models are summarized, based on the recent single-cell transcriptomics, multiomics, and spatial transcriptomics studies. While no model is perfect, it is hoped that the new insights from these models and the practical use of these models will continue to help to establish a path forward.
Collapse
Affiliation(s)
- Yuanpu Chiu
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California; Neuromedicine PhD Program, Programs in Biomedical and Biological Sciences (PIBBS), Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shangzhou Xia
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California
| | - Haowen Qiao
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Zhen Zhao
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California; Neuromedicine PhD Program, Programs in Biomedical and Biological Sciences (PIBBS), Keck School of Medicine, University of Southern California, Los Angeles, California; Neuroscience Graduate Program, University of Southern California, Los Angeles, California.
| |
Collapse
|
41
|
Seddon AR, MacArthur CP, Hampton MB, Stevens AJ. Inflammation and DNA methylation in Alzheimer's disease: mechanisms of epigenetic remodelling by immune cell oxidants in the ageing brain. Redox Rep 2024; 29:2428152. [PMID: 39579010 PMCID: PMC11587723 DOI: 10.1080/13510002.2024.2428152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease involving memory impairment, confusion, and behavioural changes. The disease is characterised by the accumulation of amyloid beta plaques and neurofibrillary tangles in the brain, which disrupt normal neuronal function. There is no known cure for Alzheimer's disease and due to increasing life expectancy, occurrence is projected to rise over the coming decades. The causes of Alzheimer's disease are multifactorial with inflammation, oxidative stress, genetic and epigenetic variation, and cerebrovascular abnormalities among the strongest contributors. We review the current literature surrounding inflammation and epigenetics in Alzheimer's disease, with a focus on how oxidants from infiltrating immune cells have the potential to alter DNA methylation profiles in the ageing brain.
Collapse
Affiliation(s)
- A. R. Seddon
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - C. P. MacArthur
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| | - M. B. Hampton
- Mātai Hāora – Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - A. J. Stevens
- Department of Pathology and Molecular Medicine, University of Otago, Wellington, New Zealand
| |
Collapse
|
42
|
Tuddenham JF, Taga M, Haage V, Marshe VS, Roostaei T, White C, Lee AJ, Fujita M, Khairallah A, Zhang Y, Green G, Hyman B, Frosch M, Hopp S, Beach TG, Serrano GE, Corboy J, Habib N, Klein HU, Soni RK, Teich AF, Hickman RA, Alcalay RN, Shneider N, Schneider J, Sims PA, Bennett DA, Olah M, Menon V, De Jager PL. A cross-disease resource of living human microglia identifies disease-enriched subsets and tool compounds recapitulating microglial states. Nat Neurosci 2024; 27:2521-2537. [PMID: 39406950 DOI: 10.1038/s41593-024-01764-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/13/2024] [Indexed: 12/06/2024]
Abstract
Human microglia play a pivotal role in neurological diseases, but we still have an incomplete understanding of microglial heterogeneity, which limits the development of targeted therapies directly modulating their state or function. Here, we use single-cell RNA sequencing to profile 215,680 live human microglia from 74 donors across diverse neurological diseases and CNS regions. We observe a central divide between oxidative and heterocyclic metabolism and identify microglial subsets associated with antigen presentation, motility and proliferation. Specific subsets are enriched in susceptibility genes for neurodegenerative diseases or the disease-associated microglial signature. We validate subtypes in situ with an RNAscope-immunofluorescence pipeline and high-dimensional MERFISH. We also leverage our dataset as a classification resource, finding that induced pluripotent stem cell model systems capture substantial in vivo heterogeneity. Finally, we identify and validate compounds that recapitulate certain subtypes in vitro, including camptothecin, which downregulates the signature of disease-enriched subtypes and upregulates a signature previously associated with Alzheimer's disease.
Collapse
Affiliation(s)
- John F Tuddenham
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Mariko Taga
- Center for Translational & 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
| | - Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Victoria S Marshe
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tina Roostaei
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles White
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Annie J Lee
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anthony Khairallah
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Gilad Green
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Sarah Hopp
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | | | | | - John Corboy
- Department of Neurology, University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora, CO, USA
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hans-Ulrich Klein
- Center for Translational & 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
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Richard A Hickman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Neil Shneider
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Eleanor and Lou Gehrig ALS Center, Columbia University Medical Center, New York, NY, USA
| | - Julie Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Marta Olah
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
43
|
Li E, Benitez C, Boggess SC, Koontz M, Rose IV, Martinez D, Draeger N, Teter OM, Samelson AJ, Pierce N, Ullian EM, Kampmann M. CRISPRi-based screens in iAssembloids to elucidate neuron-glia interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.26.538498. [PMID: 37163077 PMCID: PMC10168378 DOI: 10.1101/2023.04.26.538498] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The sheer complexity of the brain has complicated our ability to understand the cellular and molecular mechanisms underlying its function in health and disease. Genome-wide association studies have uncovered genetic variants associated with specific neurological phenotypes and diseases. In addition, single-cell transcriptomics have provided molecular descriptions of specific brain cell types and the changes they undergo during disease. Although these approaches provide a giant leap forward towards understanding how genetic variation can lead to functional changes in the brain, they do not establish molecular mechanisms. To address this need, we developed a 3D co-culture system termed iAssembloids (induced multi-lineage assembloids) that enables the rapid generation of homogenous neuron-glia spheroids. We characterize these iAssembloids with immunohistochemistry and single-cell transcriptomics and combine them with large-scale CRISPRi-based screens. In our first application, we ask how glial and neuronal cells interact to control neuronal death and survival. Our CRISPRi-based screens identified that GSK3β inhibits the protective NRF2-mediated oxidative stress response in the presence of reactive oxygen species elicited by high neuronal activity, which was not previously found in 2D monoculture neuron screens. We also apply the platform to investigate the role of APOE- ε4, a risk variant for Alzheimer's Disease, in its effect on neuronal survival. We find that APOE- ε4-expressing astrocytes may promote more neuronal activity as compared to APOE- ε3-expressing astrocytes. This platform expands the toolbox for the unbiased identification of mechanisms of cell-cell interactions in brain health and disease.
Collapse
Affiliation(s)
- Emmy Li
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Camila Benitez
- TETRAD Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Steven C. Boggess
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Mark Koontz
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Indigo V.L. Rose
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Delsy Martinez
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Nina Draeger
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Olivia M. Teter
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, USA
| | - Avi J. Samelson
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
| | - Na’im Pierce
- FirstGen Internship, Emerson Collective, USA; University of California, Berkeley, Berkeley, CA, USA
| | - Erik M. Ullian
- Department of Ophthalmology, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
44
|
Niemeyer CS, Merle L, Bubak AN, Baxter BD, Gentile Polese A, Colon-Reyes K, Vang S, Hassell JE, Bruce KD, Nagel MA, Restrepo D. Olfactory and trigeminal routes of HSV-1 CNS infection with regional microglial heterogeneity. J Virol 2024; 98:e0096824. [PMID: 39475273 PMCID: PMC11575344 DOI: 10.1128/jvi.00968-24] [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: 06/03/2024] [Accepted: 09/29/2024] [Indexed: 11/06/2024] Open
Abstract
Herpes simplex virus type 1 (HSV-1) primarily targets the oral and nasal epithelia before establishing latency in the trigeminal ganglion (TG) and other peripheral ganglia. HSV-1 can also infect and become latent in the central nervous system (CNS) independent of latency in the TGs. Recent studies suggest entry to the CNS via two distinct routes: the TG-brainstem connection and olfactory nerve; however, to date, there is no characterization of brain regions targeted during HSV-1 primary infection. Furthermore, the immune response by microglia may also contribute to the heterogeneity between different brain regions. However, the response to HSV-1 by microglia has not been characterized in a region-specific manner. This study investigated the time course of HSV-1 spread within the olfactory epithelium (OE) and CNS following intranasal inoculation and the corresponding macrophage/microglial response in a C57BL/6 mouse model. We found an apical to basal spread of HSV-1 within the OE and underlying tissue accompanied by an inflammatory response of macrophages. OE infection was followed by infection of a small subset of brain regions targeted by the TG in the brainstem and other cranial nerve nuclei, including the vagus and hypoglossal nerve. Furthermore, other brain regions were positive for HSV-1 antigens, such as the locus coeruleus (LC), raphe nucleus (RaN), and hypothalamus while sparing the hippocampus and cortex. Within each brain region, microglia activation also varied widely. These findings provide critical insights into the region-specific dissemination of HSV-1 within the CNS, elucidating potential mechanisms linking viral infection to neurological and neurodegenerative diseases.IMPORTANCEThis study shows how herpes simplex virus type 1 (HSV-1) spreads within the brain after infecting the nasal passages. Our data reveal the distinct pattern of HSV-1 through the brain during a non-encephalitic infection. Furthermore, microglial activation was also temporally and spatially specific, with some regions of the brain having sustained microglial activation even in the absence of viral antigens. Previous reports have identified specific brain regions found to be positive for HSV-1 infection; however, to date, there has not been a concise investigation of the anatomical spread of HSV-1 and the brain regions consistently vulnerable to viral entry and spread. Understanding these region-specific differences in infection and immune response is crucial because it links HSV-1 infection to potential triggers for neurological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Christy S. Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laetitia Merle
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, Dijon, France
| | - Andrew N. Bubak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - B. Dnate’ Baxter
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Arianna Gentile Polese
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine Colon-Reyes
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Sandy Vang
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James E. Hassell
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kimberley D. Bruce
- Department of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Maria A. Nagel
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
45
|
Kaji S, Berghoff SA, Spieth L, Schlaphoff L, Sasmita AO, Vitale S, Büschgens L, Kedia S, Zirngibl M, Nazarenko T, Damkou A, Hosang L, Depp C, Kamp F, Scholz P, Ewers D, Giera M, Ischebeck T, Wurst W, Wefers B, Schifferer M, Willem M, Nave KA, Haass C, Arzberger T, Jäkel S, Wirths O, Saher G, Simons M. Apolipoprotein E aggregation in microglia initiates Alzheimer's disease pathology by seeding β-amyloidosis. Immunity 2024; 57:2651-2668.e12. [PMID: 39419029 DOI: 10.1016/j.immuni.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 02/09/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
The seeded growth of pathogenic protein aggregates underlies the pathogenesis of Alzheimer's disease (AD), but how this pathological cascade is initiated is not fully understood. Sporadic AD is linked genetically to apolipoprotein E (APOE) and other genes expressed in microglia related to immune, lipid, and endocytic functions. We generated a transgenic knockin mouse expressing HaloTag-tagged APOE and optimized experimental protocols for the biochemical purification of APOE, which enabled us to identify fibrillary aggregates of APOE in mice with amyloid-β (Aβ) amyloidosis and in human AD brain autopsies. These APOE aggregates that stained positive for β sheet-binding dyes triggered Aβ amyloidosis within the endo-lysosomal system of microglia, in a process influenced by microglial lipid metabolism and the JAK/STAT signaling pathway. Taking these observations together, we propose a model for the onset of Aβ amyloidosis in AD, suggesting that the endocytic uptake and aggregation of APOE by microglia can initiate Aβ plaque formation.
Collapse
Affiliation(s)
- Seiji Kaji
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Stefan A Berghoff
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| | - Lena Spieth
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Lennart Schlaphoff
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Andrew O Sasmita
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Simona Vitale
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Luca Büschgens
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Shreeya Kedia
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martin Zirngibl
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Taisiia Nazarenko
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, Göttingen, Germany
| | - Constanze Depp
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Frits Kamp
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Patricia Scholz
- Department of Plant Biochemistry, University of Goettingen, Albrecht-von-Haller-Institute for Plant Sciences, University of Göttingen, Göttingen, Germany
| | - David Ewers
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Albinusdreef 2, 2333ZA Leiden, the Netherlands
| | - Till Ischebeck
- Department of Plant Biochemistry, University of Goettingen, Albrecht-von-Haller-Institute for Plant Sciences, University of Göttingen, Göttingen, Germany; Institute of Plant Biology and Biotechnology (IBBP), Green Biotechnology, University of Münster, Münster, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Developmental Genetics, Neuherberg, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Michael Willem
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Klaus-Armin Nave
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians University of Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians University of Munich, Munich, Germany; Department of Psychiatry and Psychotherapy, Ludwig-Maximilians University Hospital, Munich, Germany
| | - Sarah Jäkel
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Gesine Saher
- Max Planck Insitute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
46
|
Yang B, Hu S, Jiang Y, Xu L, Shu S, Zhang H. Advancements in Single-Cell RNA Sequencing Research for Neurological Diseases. Mol Neurobiol 2024; 61:8797-8819. [PMID: 38564138 DOI: 10.1007/s12035-024-04126-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Neurological diseases are a major cause of the global burden of disease. Although the mechanisms of the occurrence and development of neurological diseases are not fully clear, most of them are associated with cells mediating neuroinflammation. Yet medications and other therapeutic options to improve treatment are still very limited. Single-cell RNA sequencing (scRNA-seq), as a delightfully potent breakthrough technology, not only identifies various cell types and response states but also uncovers cell-specific gene expression changes, gene regulatory networks, intercellular communication, and cellular movement trajectories, among others, in different cell types. In this review, we describe the technology of scRNA-seq in detail and discuss and summarize the application of scRNA-seq in exploring neurological diseases, elaborating the corresponding specific mechanisms of the diseases as well as providing a reliable basis for new therapeutic approaches. Finally, we affirm that scRNA-seq promotes the development of the neuroscience field and enables us to have a deeper cellular understanding of neurological diseases in the future, which provides strong support for the treatment of neurological diseases and the improvement of patients' prognosis.
Collapse
Affiliation(s)
- Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuqi Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiru Jiang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
47
|
Fiore NT, Hayes JP, Williams SI, Moalem-Taylor G. Interleukin-35 alleviates neuropathic pain and induces an anti-inflammatory shift in spinal microglia in nerve-injured male mice. Brain Behav Immun 2024; 122:287-300. [PMID: 39097202 DOI: 10.1016/j.bbi.2024.07.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Immune cells are critical in promoting neuroinflammation and neuropathic pain and in facilitating pain resolution, depending on their inflammatory and immunoregulatory cytokine response. Interleukin (IL)-35, secreted by regulatory immune cells, is a member of the IL-12 family with a potent immunosuppressive function. In this study, we investigated the effects of IL-35 on pain behaviors, spinal microglia phenotype following peripheral nerve injury, and in vitro microglial cultures in male and female mice. Intrathecal recombinant IL-35 treatment alleviated mechanical pain hypersensitivity prominently in male mice, with only a modest effect in female mice after sciatic nerve chronic constriction injury (CCI). IL-35 treatment resulted in sex-specific microglial changes following CCI, reducing inflammatory microglial markers and upregulating anti-inflammatory markers in male mice. Spatial transcriptomic analysis revealed that IL-35 suppressed microglial complement activation in the superficial dorsal horn in male mice after CCI. Moreover, in vitro studies showed that IL-35 treatment of cultured inflammatory microglia mitigated their hypertrophied morphology, increased their cell motility, and decreased their phagocytic activity, indicating a phenotypic shift towards homeostatic microglia. Further, IL-35 altered microglial cytokines/chemokines in vitro, suppressing the release of IL-9 and monocyte-chemoattractant protein-1 and increasing IL-10 in the supernatant of male microglial cultures. Our findings indicate that treatment with IL-35 modulates spinal microglia and alleviates neuropathic pain in male mice, suggesting IL-35 as a potential sex-specific targeted immunomodulatory treatment for neuropathic pain.
Collapse
Affiliation(s)
- Nathan T Fiore
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Jessica P Hayes
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Sarah I Williams
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia
| | - Gila Moalem-Taylor
- Translational Neuroscience Facility, Department of Physiology, School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW, Australia.
| |
Collapse
|
48
|
Boon M, Akkari L. Architecture sets the path: Breast cancer subtypes differently shape the early brain metastatic niche. Cancer Cell 2024; 42:1643-1645. [PMID: 39332398 DOI: 10.1016/j.ccell.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024]
Abstract
The ability of disseminated cancer cells to colonize the brain is highly dependent on initial survival cues, often evoking early microenvironmental adaptations. In this issue of Cancer Cell, Gan et al. unveil disparate tumor architectures in early stage HER2+ breast cancer and triple-negative breast cancer brain metastases that shape stromal interactions, providing a rationale for subtype-dependent patient stratification.
Collapse
Affiliation(s)
- Menno Boon
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Leila Akkari
- Division of Tumour Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
49
|
Gan S, Macalinao DG, Shahoei SH, Tian L, Jin X, Basnet H, Bibby C, Muller JT, Atri P, Seffar E, Chatila W, Karacay A, Chanda P, Hadjantonakis AK, Schultz N, Brogi E, Bale TA, Moss NS, Murali R, Pe'er D, Massagué J. Distinct tumor architectures and microenvironments for the initiation of breast cancer metastasis in the brain. Cancer Cell 2024; 42:1693-1712.e24. [PMID: 39270646 DOI: 10.1016/j.ccell.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 12/15/2023] [Accepted: 08/20/2024] [Indexed: 09/15/2024]
Abstract
Brain metastasis, a serious complication of cancer, hinges on the initial survival, microenvironment adaptation, and outgrowth of disseminated cancer cells. To understand the early stages of brain colonization, we investigated two prevalent sources of cerebral relapse, triple-negative (TNBC) and HER2+ (HER2BC) breast cancers. Using mouse models and human tissue samples, we found that these tumor types colonize the brain, with a preference for distinctive tumor architectures, stromal interfaces, and autocrine programs. TNBC models tend to form perivascular sheaths with diffusive contact with astrocytes and microglia. In contrast, HER2BC models tend to form compact spheroids driven by autonomous tenascin C production, segregating stromal cells to the periphery. Single-cell transcriptomics of the tumor microenvironment revealed that these architectures evoke differential Alzheimer's disease-associated microglia (DAM) responses and engagement of the GAS6 receptor AXL. The spatial features of the two modes of brain colonization have relevance for leveraging the stroma to treat brain metastasis.
Collapse
Affiliation(s)
- Siting Gan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Danilo G Macalinao
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sayyed Hamed Shahoei
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lin Tian
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xin Jin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310024, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province 310024, China
| | - Harihar Basnet
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Catherine Bibby
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - James T Muller
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pranita Atri
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Evan Seffar
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Walid Chatila
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ali Karacay
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pharto Chanda
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nikolaus Schultz
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Edi Brogi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tejus A Bale
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nelson S Moss
- Department of Neurological Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rajmohan Murali
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
50
|
Zhu Y, Kohli N, Young A, Sheldon M, Coni J, Rajasekaran M, Robinson L, Chroneos R, Riley S, Guarnieri JW, Jose J, Patel N, Wallace DC, Li S, Lee H, Mach RH, McManus MJ. PET Imaging with [ 18F]ROStrace Detects Oxidative Stress and Predicts Parkinson's Disease Progression in Mice. Antioxidants (Basel) 2024; 13:1226. [PMID: 39456479 PMCID: PMC11504722 DOI: 10.3390/antiox13101226] [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: 08/20/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although the precise molecular mechanisms responsible for neuronal death and motor dysfunction in late-onset Parkinson's disease (PD) are unknown, evidence suggests that mitochondrial dysfunction and neuroinflammation occur early, leading to a collective increase in reactive oxygen species (ROS) production and oxidative stress. However, the lack of methods for tracking oxidative stress in the living brain has precluded its use as a potential biomarker. The goal of the current study is to address this need through the evaluation of the first superoxide (O2•-)-sensitive radioactive tracer, [18F]ROStrace, in a model of late-onset PD. To achieve this goal, MitoPark mice with a dopaminergic (DA) neuron-specific deletion of transcription factor A mitochondrial (Tfam) were imaged with [18F]ROStrace from the prodromal phase to the end-stage of PD-like disease. Our data demonstrate [18F]ROStrace was sensitive to increased oxidative stress during the early stages of PD-like pathology in MitoPark mice, which persisted throughout the disease course. Similarly to PD patients, MitoPark males had the most severe parkinsonian symptoms and metabolic impairment. [18F]ROStrace retention was also highest in MitoPark males, suggesting oxidative stress as a potential mechanism underlying the male sex bias of PD. Furthermore, [18F]ROStrace may provide a method to identify patients at risk of Parkinson's before irreparable neurodegeneration occurs and enhance clinical trial design by identifying patients most likely to benefit from antioxidant therapies.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neha Kohli
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anthony Young
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malkah Sheldon
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jani Coni
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Meera Rajasekaran
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lozen Robinson
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rea Chroneos
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shaipreeah Riley
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Joshua Jose
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nisha Patel
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shihong Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meagan J. McManus
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|