1
|
Li S, Guo Y, Zhou X, Li C, Hong Y, Li M, Zhang Q, Ning B, Jiang Y. Multifunctional albumin-based hydrogel/microglia composites enhancing the therapeutic potential of neonatal microglia in complex spinal cord injuries and sealing dural rupture. Biomaterials 2025; 321:123327. [PMID: 40220568 DOI: 10.1016/j.biomaterials.2025.123327] [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: 10/06/2024] [Revised: 02/17/2025] [Accepted: 04/06/2025] [Indexed: 04/14/2025]
Abstract
Treatment for spinal cord injuries (SCIs) remains largely ineffective, with scar formation and neural degeneration being major barriers to functional recovery. Neonatal microglia have shown potential in reducing scar formation and promoting axonal regrowth. However, cell viability and retention at the injury site are often suboptimal. The hostile post-SCI inflammatory microenvironment leads to poor cell survival and the dural damage that is frequently associated with SCIs results in cell loss. To address these challenges, we have developed an albumin-based hydrogel. This hydrogel creates a favorable microenvironment for the encapsulated cells, mimicking the extracellular matrix and enhancing the viability of the transplanted cells. In vivo studies demonstrate its efficacy in preventing scar formation, promoting axonal regeneration, and sealing the dura. Importantly, this hydrogel leverages albumin, a natural polymer in the body, and is synthesized through a simple process, making it highly feasible for clinical translation. In summary, this albumin hydrogel is a valuable delivery vehicle that enhances the therapeutic potential of neonatal microglia in treating SCIs, particularly those involving dural rupture.
Collapse
Affiliation(s)
- Shang Li
- Jinan Central Hospital, Shandong University, Jinan, Shandong, 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China
| | - Yijian Guo
- Jinan Central Hospital, Shandong University, Jinan, Shandong, 250013, China
| | - Xiaoyu Zhou
- Department of Neurosurgery, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, 250033, China; Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Can Li
- Department of Neurosurgery, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, 250033, China; Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Yatian Hong
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China
| | - Mingxin Li
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China
| | - Qingchen Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, Jinan, Shandong, 250013, China; Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250013, China.
| | - Yanyan Jiang
- Department of Neurosurgery, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, 250033, China; Key Laboratory for Liquid-Solid Structural Evolution and Processing of Materials (Ministry of Education), School of Materials Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| |
Collapse
|
2
|
Sun S, Tian R, Alford A, Yin D, Shi R. An analog of phenelzine demonstrates effective acrolein scavenging and neuroprotection without monoamine oxidase inhibition in a rat SCI model. Neuroscience 2025; 574:54-64. [PMID: 40189130 DOI: 10.1016/j.neuroscience.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Oxidative stress is widely recognized as a critical factor in the functional deficits after spinal cord injury (SCI). Oxidative stress and lipid peroxidation-derived aldehydes such as acrolein are known to play a key role in SCI pathology and have therefore emerged as valuable therapeutic targets. This study introduces a novel phenelzine analogue (PhzA), designed to retain the acrolein scavenging capability of phenelzine (Phz) while removing its undesirable monoamine oxidase (MAO) inhibition effects through structure-based modification. Using a rat model of contusion SCI, we showed that PhzA significantly reduced acrolein levels in both the acute and chronic stages of SCI with minimal MAO inhibition. In addition, PhzA reduced excessive microglial and astrocytic activation, dampening inflammation and gliosis. Furthermore, PhzA-treated rats exhibited significant improvements in motor function and reduction in mechanical hypersensitivity for up to 28 days post-injury compared to untreated rats. These findings further underscore the crucial role of aldehydes in SCI pathology and strengthen the notion that acrolein could serve as an effective therapeutic target for mitigating post-SCI neurodegeneration. These results also indicate that the expansion of acrolein-scavenging drug discovery through structure-based modification of existing repurposed drugs, such as with Phz, is a viable strategy with the benefit of a likely accelerated path towards clinical application. This effort may also benefit a range of neuronal diseases and injuries beyond SCI where acrolein is implicated, advancing the health of millions of patients.
Collapse
Affiliation(s)
- Siyuan Sun
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA
| | - Ran Tian
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA
| | - Anna Alford
- Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA
| | - David Yin
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; West Lafayette Junior/Senior High School, West Lafayette, IN, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine Purdue University, West Lafayette, IN, USA; Center for Paralysis Research, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, College of Engineering, Purdue University, West Lafayette, IN, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
3
|
Wang Y, Jia X, Zhang Y, Shi H, Sun Y, Liu Y. Hydrogen Sulfide Modulates Microglial Polarization and Remodels the Injury Microenvironment to Promote Functional Recovery After Spinal Cord Injury. CNS Neurosci Ther 2025; 31:e70431. [PMID: 40365715 PMCID: PMC12076064 DOI: 10.1111/cns.70431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
AIMS Spinal cord injury (SCI) disrupts tissue homeostasis, leading to persistent neuroinflammation and scar formation that severely impedes functional recovery. Current therapeutic approaches are insufficient to address these challenges. In this study, we investigated whether exogenous hydrogen sulfide (H2S) can modulate neuroinflammatory responses and remodel the injury microenvironment to promote tissue repair and restore motor function following SCI. METHODS T10 crush SCI was induced in mice, followed by daily intraperitoneal administration of the H2S donor anethole trithione (ADT). Immunofluorescence staining, tissue clearing, western blotting, and behavioral assessments were performed to evaluate scar formation, vascular regeneration, neuronal survival, and motor function. RESULTS ADT-based H2S therapy significantly promoted wound healing, inhibited scar formation, enhanced vascular regeneration, and protected residual neurons and axons from secondary injury. Mechanistically, H2S suppressed microglial proliferation and activation, promoting their polarization toward an anti-inflammatory phenotype and alleviating neuroinflammation. Consequently, motor function recovery was markedly improved. CONCLUSION H2S modulates microglial activation and mitigates neuroinflammation, establishing a permissive microenvironment for SCI repair and significantly enhancing motor function recovery. Given ADT's established clinical safety and its effective gasotransmitter properties, our findings underscore its immediate translational potential for treating SCI.
Collapse
Affiliation(s)
- Yu Wang
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Institute of Neuroscience, Soochow University, Departments of Rehabilitation Medicine and Neurology, The Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xinyi Jia
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Institute of Neuroscience, Soochow University, Departments of Rehabilitation Medicine and Neurology, The Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yuqi Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Haibin Shi
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, and Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education InstitutionsSoochow UniversitySuzhouChina
| | - Yuhui Sun
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Institute of Neuroscience, Soochow University, Departments of Rehabilitation Medicine and Neurology, The Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yaobo Liu
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, Institute of Neuroscience, Soochow University, Departments of Rehabilitation Medicine and Neurology, The Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
- Co‐Innovation Center of Neuroregeneration, Nantong UniversityNantongChina
| |
Collapse
|
4
|
Cellini BR, Edachola SV, Faw TD, Cigliola V. Blueprints for healing: central nervous system regeneration in zebrafish and neonatal mice. BMC Biol 2025; 23:115. [PMID: 40307837 PMCID: PMC12044871 DOI: 10.1186/s12915-025-02203-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
In adult mammals, including humans, neurons, and axons in the brain and spinal cord are inherently incapable of regenerating after injury. Studies of animals with innate capacity for regeneration are providing valuable insights into the mechanisms driving tissue healing. The aim of this review is to summarize recent data on regeneration mechanisms in the brain and spinal cord of zebrafish and neonatal mice. We infer that elucidating these mechanisms and understanding how and why they are lost in adult mammals will contribute to the development of strategies to promote central nervous system regeneration.
Collapse
Affiliation(s)
- Brianna R Cellini
- Department of Psychology and Neuroscience, Duke University, Durham, NC, 27710, USA
| | | | - Timothy D Faw
- Department of Orthopaedic Surgery, Duke University, Durham, NC, 27710, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, 27710, USA
| | - Valentina Cigliola
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
5
|
Liu Y, Yao F, Li Z, Jiang Y, Li J, Yu S, Hu X, Ouyang F, Zheng M, Cheng L, Jing J. Dynamic phosphorylation of Fascin-1 orchestrates microglial phagocytosis and neurological recovery after spinal cord injury. J Neuroinflammation 2025; 22:121. [PMID: 40281563 PMCID: PMC12032802 DOI: 10.1186/s12974-025-03445-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
The persistence of myelin debris after spinal cord injury (SCI) constitutes a formidable barrier to axonal regeneration, remyelination, and functional recovery by initiating inflammatory cascades. Microglia, known for their superior phagocytic and degradative capabilities, are crucial in clearing myelin debris. Yet, the molecular mechanisms governing their function remain elusive. Our previous research has identified a sustained upregulation of Fascin-1, an actin-binding protein essential for phagocytosis, in Cx3cr1+ microglia after SCI. Here, we reveal that ablation of microglial Fascin-1 exacerbates neuronal loss and hampers motor recovery after SCI, correlating with diminished microglial phagocytic activity in Cx3cr1cre+/-;Fascin-1fl/fl mice. We demonstrated that dysregulated Fascin-1 phosphorylation impairs microglial phagocytosis, linked to the upstream Mas1/Protein kinase C gamma (PKCγ) axis. Pharmacologic activation of the Mas1/PKC axis to drive Fascin-1 phosphorylation in microglia restores phagocytic function, thereby alleviating neuronal loss and facilitating neurological recovery after SCI. Our findings underscore the critical role of Fascin-1 phosphorylation in microglial phagocytosis and highlight the Mas1/PKCγ axis as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yanchang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyu Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianjian Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuisheng Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangru Ouyang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
6
|
Jiang Q, Xue S, Pan X, Yu T, Wei X, Li L, Qi C, Shi W, Ren Z, Hu D, Fu H. Differential changes in the microglial transcriptome between neonatal and adult mice after spinal cord injury. Sci Rep 2025; 15:13708. [PMID: 40258965 PMCID: PMC12012053 DOI: 10.1038/s41598-025-98429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/11/2025] [Indexed: 04/23/2025] Open
Abstract
Spinal cord injury (SCI) remains a significant therapeutic challenge, lacking effective treatment options. Related studies have found that neonatal microglia are more effective than adult microglia in promoting the recovery of SCI, but the reason why neonatal, not adult, microglia are more conducive to SCI recovery is not clear, the differences of gene expression and pathways between them are still worth exploring. Therefore, we examined changes in the microglial transcriptome after SCI in neonatal and adult mice. We identified hub genes or pathways that exhibited significant differential expression between the two groups. Four Gene sets were established for further analysis, named Gene set 1, Gene set 2, Gene set 3, Gene set 4, respectively. GO analysis revealed enrichment in categories critical for injury repair, including DNA metabolism, replication, recombination, meiotic cell cycle progression, regulation of cell-cell adhesion, megakaryocyte and endothelial development, modulation of the neuroinflammatory response, endocytosis, and regulation of cytokine production and cell migration. KEGG analysis revealed enrichment in pathways critical for various cellular processes, including the p53, TNF, PI3K-AKT, PPAR and B cell receptor signaling pathway, axon guidance, cytokine-cytokine receptor interaction. PPI and TF-hub gene-microRNA networks were constructed to elucidate the underlying gene regulatory mechanisms. Additionally, drug prediction was performed to identify potential therapeutic candidates. Finally, 11 hub genes (Chek1, RRM2, Lyve1, Mboat1, Clec4a3, Ccnd1, Cdk6, Zeb1, Igf1, Pparg, and Cd163) were selected from four Gene sets for further validation using qRT-PCR. We identified candidate genes and pathways involved in microglial transcriptome heterogeneity after SCI in neonatal and adult mice. These findings provide valuable insights into potential therapeutic targets for neonatal microglia in the treatment of SCI.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Qingdao Medical College of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Shiyuan Xue
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Qingdao Medical College of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Xiaojing Pan
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China
| | - Tengbo Yu
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Xinyi Wei
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Qingdao Medical College of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Liping Li
- Department of Bone Surgery, Qingdao Central Hospital, Qingdao, 266000, China
| | - Chao Qi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Qingdao Medical College of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Zhongkai Ren
- Department of Orthopedic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Die Hu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, Qingdao, 266071, China.
| | - Haitao Fu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
7
|
Weber RZ, Achón Buil B, Rentsch NH, Bosworth A, Zhang M, Kisler K, Tackenberg C, Rust R. A molecular brain atlas reveals cellular shifts during the repair phase of stroke. J Neuroinflammation 2025; 22:112. [PMID: 40251566 PMCID: PMC12008922 DOI: 10.1186/s12974-025-03437-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/02/2025] [Indexed: 04/20/2025] Open
Abstract
Ischemic stroke triggers a cascade of pathological events that affect multiple cell types and often lead to incomplete functional recovery. Despite advances in single-cell technologies, the molecular and cellular responses that contribute to long-term post-stroke impairment remain poorly understood. To gain better insight into the underlying mechanisms, we generated a single-cell transcriptomic atlas from distinct brain regions using a mouse model of permanent focal ischemia at one month post-injury. Our findings reveal cell- and region-specific changes within the stroke-injured and peri-infarct brain tissue. For instance, GABAergic and glutamatergic neurons exhibited upregulated genes in signaling pathways involved in axon guidance and synaptic plasticity, and downregulated pathways associated with aerobic metabolism. Using cell-cell communication analysis, we identified increased strength in predicted interactions within stroke tissue among both neural and non-neural cells via signaling pathways such as those involving collagen, protein tyrosine phosphatase receptor, neuronal growth regulator, laminin, and several cell adhesion molecules. Furthermore, we found a strong correlation between mouse transcriptome responses after stroke and those observed in human nonfatal brain stroke lesions. Common molecular features were linked to inflammatory responses, extracellular matrix organization, and angiogenesis. Our findings provide a detailed resource for advancing our molecular understanding of stroke pathology and for discovering therapeutic targets in the repair phase of stroke recovery.
Collapse
Affiliation(s)
- Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, 8057, Switzerland
| | - Beatriz Achón Buil
- Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, 8057, Switzerland
| | - Nora H Rentsch
- Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, 8057, Switzerland
| | - Allison Bosworth
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Mingzi Zhang
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, 8952, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, 8057, Switzerland
| | - Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, 90033, USA.
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
8
|
Sun W, Dion E, Laredo F, Okonak A, Sepeda JA, Haykal E, Zhou M, El-Hodiri HM, Fischer AJ, Silver J, Peng J, Sas A, Tedeschi A. In vivo programming of adult pericytes aids axon regeneration by providing cellular bridges for SCI repair. Mol Ther 2025:S1525-0016(25)00294-1. [PMID: 40253585 DOI: 10.1016/j.ymthe.2025.04.020] [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: 06/21/2024] [Revised: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Pericytes are contractile cells of the microcirculation that participate in wound healing after spinal cord injury (SCI). Thus far, the extent to which pericytes cause or contribute to axon growth and regeneration failure after SCI remains controversial. Here, we found that SCI leads to profound changes in vasculature architecture and pericyte coverage. We demonstrated that pericytes constrain sensory axons on their surface, causing detrimental structural and functional changes in adult dorsal root ganglion neurons that contribute to axon regeneration failure after SCI. Perhaps more excitingly, we discovered that in vivo programming of adult pericytes via local administration of platelet-derived growth factor BB (PDGF-BB) effectively promotes axon regeneration and recovery of hindlimb function by contributing to the formation of cellular bridges that span the lesion. Ultrastructural analysis showed that PDGF-BB induced fibronectin fibril alignment and extension, effectively converting adult pericytes into a permissive substrate for axon growth. In addition, PDGF-BB localized delivery positively affects the physical and chemical nature of the lesion environment, thereby creating more favorable conditions for SCI repair. Thus, therapeutic manipulation rather than wholesale ablation of pericytes can be exploited to prime axon regeneration and SCI repair.
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Elliot Dion
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Fabio Laredo
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; University of Nottingham, Nottingham NG7 2QL, UK
| | - Allyson Okonak
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jesse A Sepeda
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Esraa Haykal
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Min Zhou
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem M El-Hodiri
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Juan Peng
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Sas
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
9
|
Godeanu S, Cătălin B. The Complementary Role of Morphology in Understanding Microglial Functional Heterogeneity. Int J Mol Sci 2025; 26:3811. [PMID: 40332469 PMCID: PMC12027755 DOI: 10.3390/ijms26083811] [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: 03/13/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
A search of the PubMed database for publications on microglia reveals an intriguing shift in scientific interest over time. Dividing microglia into categories such as "resting" and "activated" or M1 versus M2 is nowadays obsolete, with the current research focusing on unraveling microglial heterogeneity. The onset of transcriptomics, especially single-cell RNA sequencing (scRNA-seq), has profoundly reshaped our understanding of microglia heterogeneity. Conversely, microglia morphology analysis can offer important insights regarding their activation state or involvement in tissue responses. This review explores microglial heterogeneity under homeostatic conditions, developmental stages, and disease states, with a focus on integrating transcriptomic data with morphological analysis. Beyond the core gene expression profile, regional differences are observed with cerebellar microglia exhibiting a uniquely immune-vigilant profile. During development, microglia express homeostatic genes before birth, yet the bushy appearance is a characteristic that appears later on. In neurodegeneration, microglia alternate between proinflammatory and neuroprotective roles, influenced by regional factors and disease onset. Understanding these structural adaptations may help identify specific microglial subpopulations for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sânziana Godeanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, University of Saarland, 66421 Homburg, Germany
| |
Collapse
|
10
|
Shen Z, Feng B, Lim WL, Woo T, Liu Y, Vicenzi S, Wang J, Kwon BK, Zou Y. Astrocytic Ryk signaling coordinates scarring and wound healing after spinal cord injury. Proc Natl Acad Sci U S A 2025; 122:e2417400122. [PMID: 40208942 PMCID: PMC12012454 DOI: 10.1073/pnas.2417400122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 04/12/2025] Open
Abstract
Wound healing after spinal cord injury involves highly coordinated interactions among multiple cell types, which are poorly understood. Astrocytes play a central role in creating a border against the non-neural lesion core. To do so, astrocytes undergo dramatic morphological changes by first thickening and elongating their processes and then overlapping them to form a physical barrier. We show here that the expression of a cell-surface receptor, Ryk, is induced in astrocytes after injury in both rodent and human spinal cords. Astrocyte-specific knockout of Ryk dramatically elongated the reactive astrocytes, accelerated the formation of the border, and reduced the size of the scar. Astrocyte-specific knockout of Ryk also accelerated the injury responses of multiple cell types. Single-cell transcriptomics analyses revealed a broad range of changes in cell signaling among astrocytes, microglia, fibroblasts, and endothelial cells after astrocyte-specific Ryk knockout, suggesting that Ryk not only regulates injury responses of astrocytes but may also regulate signals emanating from astrocytes and coordinate the responses of these cell types. The elongation of astrocyte processes is mediated by NrCAM, a cell adhesion molecule induced by astrocyte-specific conditional knockout of Ryk after spinal cord injury. Our findings suggest that Ryk is a promising therapeutic target to accelerate wound healing, promote neuronal survival, and enhance functional recovery.
Collapse
Affiliation(s)
- Zhe Shen
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Bo Feng
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Wei Ling Lim
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Timothy Woo
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Yanlin Liu
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Silvia Vicenzi
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Jingyi Wang
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| | - Brian K. Kwon
- Department of Orthopaedics, International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BCV5Z 1M9, Canada
| | - Yimin Zou
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
11
|
Cai Y, Lin Z, Shen X, Li M, Xing L, Yang T, Chen G. Effect of microglial Pd1 on glial scar formation after spinal cord injury in mice. J Biol Chem 2025; 301:108489. [PMID: 40209954 DOI: 10.1016/j.jbc.2025.108489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The cross talk between microglia and astrocytes following spinal cord injury (SCI) greatly decides the prognosis. However, a comprehensive understanding of the molecular mechanisms by which microglia regulate astrocytic activity post-SCI is lacking. Programmed cell death protein 1 (Pdcd1, Pd1) plays a crucial role in modulating immune responses by exerting suppressive effects on microglia and peripheral immune cells within the central nervous system. Previous studies have shown the involvement of Pd1 in the pathogenesis of SCI; however, the role of microglial Pd1 in astrocytic activation and the following glial scar formation remains elusive. Here, we demonstrated that the pharmacological depletion of microglia using minocycline decreased the expression of tumor necrosis factor-alpha and interleukin-6 while concurrently increasing the expression of interleukin-10 following SCI, thereby facilitating motor function recovery in mice. We observed an increase in Pd1 expression in the injured spinal cord after SCI, with precise localization of Pd1 within microglia. Based on Pd1 knockout (KO) mice, we further revealed that Pd1 deficiency disrupted glial scar formation, leading to increased inflammation, impeded nerve regeneration, enlarged tissue damage, and compromised functional recovery following SCI. In vitro study showed that siRNA-mediated inhibition of Pd1 in microglia followed by lipopolysaccharide treatment significantly inhibited astrocyte migration and upregulated the secretion of tumor necrosis factor-alpha and CXCL9 from microglia, indicating that microglial Pd1 regulates glial scar formation through modulating the inflammatory microenvironment. Our study gains a new mechanistic insight into how microglial Pd1 decides the fate of SCI and promotes microglial Pd1 as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Zhihao Lin
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China
| | - Xin Shen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Ming Li
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
12
|
Hellenbrand DJ, Lee JS, Mickelson EJ, Baer MC, Ott EL, Martinson NR, Ceelen MR, Hilger KH, Nielsen BE, Jacobs AN, Mishra RR, Hurley SA, Murphy WL, Hanna AS. Mineral coated microparticles delivering Interleukin-4, Interleukin-10, and Interleukin-13 reduce inflammation and improve function after spinal cord injury in a rat. Exp Neurol 2025; 386:115179. [PMID: 39914642 DOI: 10.1016/j.expneurol.2025.115179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
After spinal cord injury (SCI) there is excessive inflammation and extensive infiltration of immune cells that leads to additional neural damage. Interleukin (IL)-4, IL-10, and IL-13 are anti-inflammatories that have been shown to reduce several pro-inflammatory species, alter macrophage state, and provide neuroprotection. However, these anti-inflammatories have a short half-life, do not cross the blood-spinal cord barrier, and large systemic doses of ant-inflammatory cytokines can cause increased susceptibility to infections. In this study, we used mineral coated microparticles (MCMs) to bind, stabilize and deliver biologically active IL-4, IL-10, and IL-13 in a sustained manner directly to the injury site. Rats with a T10 SCI were given an intraspinal injection of cytokine-loaded MCMs 6 h post-injury. Testing of 27 cytokine/chemokine levels 24 h post-injury demonstrated that MCMs delivering IL-4, IL-10, and IL-13 significantly reduced inflammation (P < 0.0001). Rats treated with MCMs+(IL-4, IL-10, IL-13) had significantly higher Basso-Beattie-Bresnahan locomotor rating scores (P = 0.0021), Ladder Rung Test scores (P = 0.0021), and significantly longer latency threshold with the Hargreaves Test (P = 0.0123), compared to Injured Controls. Analyses of post-fixed spinal cords revealed significantly less spinal cord atrophy (P = 0.0344) in rats treated with MCMs+(IL-4, IL-10, IL-13), and diffusion tensor imaging tractography revealed significantly more tracts spanning the injury site (P = 0.0025) in rats treated with MCMs+(IL-4, IL-10, IL-13) compared to Injured Controls. In conclusion, MCMs delivering IL-4, IL-10, and IL-13 significantly reduced inflammation post-SCI, resulting in significantly less spinal cord damage and a significant improvement in hind limb function.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ethan J Mickelson
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Matthew C Baer
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Emily L Ott
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Natalie R Martinson
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Matthew R Ceelen
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Keegan H Hilger
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Brooke E Nielsen
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Alison N Jacobs
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raveena R Mishra
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Samuel A Hurley
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Forward BIO Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Amgad S Hanna
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
13
|
Diep YN, Park HJ, Zhu X, Nam JH, Shim JK, Chang JH, Nguyen DD, Kim I, Jo DG, Lee LP, Yun M, Cho H. A Neuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) for Evaluating Astrocytic Scar Formation and Microgliosis in Glioblastoma Microenvironment. Adv Healthc Mater 2025; 14:e2404821. [PMID: 40072333 DOI: 10.1002/adhm.202404821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/28/2025] [Indexed: 04/26/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, characterized by its heterogeneity in cellular components, including reactive astrocytes and microglia. Since neuroimmune responses like astrogliosis and microgliosis gain recognition as vital factors in brain tumor progression, there is a growing need for clinically relevant models that assess the interactions between astrocytes, microglia, and GBM. Here, a NEuroimmune-Oncology Microphysiological Analysis Platform (NEO-MAP) is presented as a "new map" to observe astrocytic scar formation and microgliosis in response to GBM. NEO-MAP based on pathophysiological principles is designed to replicate the GBM-glia interactions, multi-phenotypic microglia activities, scar-forming astrocytes with chondroitin sulfate proteoglycans (CSPGs) in the extracellular matrix, and the biophysical characteristics of the astrocytic scar barrier. The NEO-MAP reveals that inhibiting mTORC2 in GBM promotes the proinflammatory transformation of astrocytes and enhanced astrocytic scar formation. Astrocytes that form scars prompted microglia to change from the M2 to M1 phenotype, enhancing chemotherapy sensitivity. Tissues from GBM patients show a significant correlation between reduced mTORC2 activity and increased astrogliosis, alongside a decrease in M2-polarized microgliosis, aligning with the NEO-MAP findings. Overall, the NEO-MAP is foreseen as a clinically significant tool for exploring tumor-glia interactions, opening avenues for drug development aimed at the tumor microenvironment.
Collapse
Affiliation(s)
- Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee Jung Park
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Xiaohui Zhu
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jin Ho Nam
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Dang Du Nguyen
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inki Kim
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Dong-Gyu Jo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Luke P Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA
| | - Mijin Yun
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
14
|
Valdearcos M, McGrath ER, Brown Mayfield SM, Jacuinde MG, Folick A, Cheang RT, Li R, Bachor TP, Lippert RN, Xu AW, Koliwad SK. Microglia mediate the early-life programming of adult glucose control. Cell Rep 2025; 44:115409. [PMID: 40085644 DOI: 10.1016/j.celrep.2025.115409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/28/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
Glucose homeostasis is, in part, nutritionally programmed during early neonatal life, a critical window for synapse formation between hypothalamic glucoregulatory centers. Although microglia prune synapses throughout the brain, their role in refining hypothalamic glucoregulatory circuits remains unclear. Here, we show that the phagocytic activity of microglia in the mediobasal hypothalamus (MBH) is induced following birth, regresses upon weaning from maternal milk, and is exacerbated by feeding dams a high-fat diet while lactating. In addition to actively engulfing synapses, microglia are critical for refining perineuronal nets (PNNs) within the neonatal MBH. Remarkably, transiently depleting microglia before weaning (postnatal day [P]6-16) but not afterward (P21-31) induces glucose intolerance in adulthood due to impaired insulin responsiveness, which we link to PNN overabundance and reduced synaptic connectivity between hypothalamic glucoregulatory neurons and the pancreatic β cell compartment. Thus, microglia facilitate early-life synaptic plasticity in the MBH, including PNN refinement, to program hypothalamic circuits regulating adult glucose homeostasis.
Collapse
Affiliation(s)
- Martin Valdearcos
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Emily R McGrath
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | | | - Melissa G Jacuinde
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew Folick
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Rachel T Cheang
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Ruoyu Li
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Tomas P Bachor
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Rachel N Lippert
- German Institute of Human Nutrition Potsdam Rehbrücke, Potsdam, Germany; German Center for Diabetes Research, Neuherberg, Germany; Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Allison W Xu
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Suneil K Koliwad
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
15
|
Ye J, Shan F, Xu X, Liang C, Zhang N, Hu H, Li J, Ouyang F, Wang J, Zhao Y, Ma Z, Meng C, Li Z, Yu S, Jing J, Zheng M. Centripetal migration and prolonged retention of microglia promotes spinal cord injury repair. J Neuroinflammation 2025; 22:77. [PMID: 40075472 PMCID: PMC11905688 DOI: 10.1186/s12974-025-03411-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Recent studies have confirmed the critical role of neonatal microglia in wound healing and axonal regeneration following spinal cord injury (SCI). However, the limited migration of microglia to the center of adult lesion may significantly impede their potential benefits. METHODS We established a model of microglial centripetal migration and prolonged retention in C57BL/6J and transgenic mice by injecting exogenous C-X3-C motif chemokine ligand 1 (CX3CL1) and macrophage colony-stimulating factor (M-CSF) directly into the lesion site post-SCI. Wound healing and axonal preservation/regrowth was assessed anatomically, and kinematics analysis was conducted to determine the recovery of locomotor function. RESULTS We identified decreased expression and perilesional distribution of CX3CL1 as the primary reason for the limited centripetal migration of microglia. In situ injection of CX3CL1 into the lesion core promoted microglial centripetal migration, but alone did not improve functional recovery. Nevertheless, a combinational administration of CX3CL1 and M-CSF fostered both centripetal migration and prolonged retention of microglia, thereby effectively displacing blood-derived macrophage infiltration and optimizing wound healing and axonal preservation/regrowth after SCI. Notably, the beneficial effects of CX3CL1 and M-CSF co-administration were specifically blocked in C-X3-C motif chemokine receptor 1 (CX3CR1)-deficient mice. These phenomena may be related to the increase in spleen tyrosine kinase (SYK) levels, which boosts centripetal microglial phagocytosis. CONCLUSION Our study uncovers the criticality of microglial location and abundance in orchestrating SCI repair, highlighting centripetal microglial dynamics as valuable targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jianan Ye
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fangli Shan
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Xinzhong Xu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Chao Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ningyuan Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hao Hu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianjian Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fangru Ouyang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jingwen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuanzhe Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhida Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Congpeng Meng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ziyu Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Shuisheng Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Juehua Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Meige Zheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
16
|
Smith AN, Nagrabski S, Baker L, Kramer AH, Sharp DJ, Byrnes KR. Fidgetin-like 2 knockdown increases acute neuroinflammation and improves recovery in a rat model of spinal cord injury. J Neuroinflammation 2025; 22:73. [PMID: 40065364 PMCID: PMC11895163 DOI: 10.1186/s12974-025-03344-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/10/2025] [Indexed: 03/14/2025] Open
Abstract
Spinal cord injury (SCI) can cause permanent dysfunction proceeding from multifaceted neuroinflammatory processes that contribute to damage and repair. Fidgetin-like 2 (FL2), a microtubule-severing enzyme that negatively regulates axon growth, microglial functions, and wound healing, has emerged as a potential therapeutic target for central nervous system injuries and neuroinflammation. To test the hypothesis that FL2 knockdown increases acute neuroinflammation and improves recovery after SCI, we examined the effects of nanoparticle-encapsulated FL2 siRNA treatment after a moderate contusion SCI in rats. SCI significantly increased FL2 expression in the lesion site and rostral to the lesion 1 day post-injury (dpi). A single treatment of FL2 siRNA after injury led to modestly improved locomotor recovery consistent with the preservation of corticospinal tract function, accompanied by reduced inflammation and increased presence of oligodendrocytes. In determining the acute effects of treatment, RNA sequencing and gene set enrichment analyses revealed that FL2 siRNA modulates early cellular responses, including chemokine signaling, both pro- and anti-inflammatory immune reactions, and neurotransmitter signaling pathways at 1, 4, and 7 dpi. Follow-up analyses at 4 dpi using dual in situ hybridization and immunohistochemistry demonstrated that SCI increased FL2 mRNA and that FL2 was colocalized with microglia/macrophages. FL2 downregulation resulted in a marked accumulation of microglia at the lesion site, accompanied by increased inflammatory markers (IL-1β, TGF-β1, and CD68). The results suggest SCI induces an increase in FL2 expression that undermines acute inflammatory responses as well as spinal cord integrity and growth. Overall, our study suggests that targeting FL2 holds promise as a therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Austin N Smith
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Samantha Nagrabski
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | | - David J Sharp
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kimberly R Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
17
|
Ding Y. Histone deacetylases: the critical enzymes for microglial activation involved in neuropathic pain. Front Pharmacol 2025; 16:1515787. [PMID: 40115267 PMCID: PMC11922887 DOI: 10.3389/fphar.2025.1515787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/23/2025] Open
Abstract
Neuropathic pain is a common health problem in clinical practice that can be caused by many different factors, including infection, ischemia, trauma, diabetes mellitus, nerve compression, autoimmune disorders, cancer, trigeminal neuralgia, and abuse of certain drugs. This type of pain can persistently affect patients for a long time, even after the rehabilitation of their damaged tissues. Researchers have identified the crucial role of microglial activation in the pathogenesis of neuropathic pain. Furthermore, emerging evidence has shown that the expression and/or activities of different histone deacetylases (HDACs) can modulate microglial function and neuropathic pain. In this review, we will summarize and discuss the functions and mechanisms of HDACs in microglial activation and neuropathic pain development. Additionally, we will also list the emerging HDAC inhibitors or activators that may contribute to therapeutic advancement in alleviating neuropathic pain.
Collapse
Affiliation(s)
- Yi Ding
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
18
|
Xian Y, Liu J, Dai M, Zhang W, He M, Wei Z, Jiang Y, Le S, Lin Z, Tang S, Zhou Y, Dong L, Liang J, Zhang J, Wang L. Microglia Promote Lymphangiogenesis Around the Spinal Cord Through VEGF-C/VEGFR3-Dependent Autophagy and Polarization After Acute Spinal Cord Injury. Mol Neurobiol 2025; 62:2740-2755. [PMID: 39158788 DOI: 10.1007/s12035-024-04437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Reducing secondary injury is a key focus in the field of spinal cord injury (SCI). Recent studies have revealed the role of lymphangiogenesis in reducing secondary damage to central nerve. However, the mechanism of lymphangiogenesis is not yet clear. Macrophages have been shown to play an important role in peripheral tissue lymphangiogenesis. Microglia is believed to play a role similar to macrophages in the central nervous system (CNS); we hypothesized that there was a close relationship between microglia and central nerve system lymphangiogenesis. Herein, we used an in vivo model of SCI to explored the relationship between microglia and spinal cord lymphangiogenesis and further investigated the polarization of microglia and its role in promoting spinal cord lymphangiogenesis by a series of in vitro experiments. The current study elucidated for the first time the relationship between microglia and lymphangiogenesis around the spinal cord after SCI. Classical activated (M1) microglia can promote lymphangiogenesis by secreting VEGF-C which further increases polarization and secretion of lymphatic growth factor by activating VEGFR3. The VEGF-C/VEGFR3 pathway activation downregulates microglia autophagy, thereby regulating the microglia phenotype. These results indicate that M1 microglia promote lymphangiogenesis after SCI, and activated VEGF-C/VEGFR3 signaling promotes M1 microglia polarization by inhibiting autophagy, thereby facilitates lymphangiogenesis.
Collapse
Affiliation(s)
- Yeyang Xian
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Liu
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Mengxuan Dai
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Wensheng Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Minye He
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhengnong Wei
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yutao Jiang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shiyong Le
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Zhuoang Lin
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Shuai Tang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Yunfei Zhou
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Liming Dong
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jinzheng Liang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China
| | - Jie Zhang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| | - Liang Wang
- Tianhe District, Guangdong Province, The Third Affiliated Hospital, Southern Medical University, Zhongshandadao West 183, Guangzhou City, 510000, China.
| |
Collapse
|
19
|
Lin Y, Lang H, Gao P, Miao X, Guo Q, Hao Y, Ai T, Li J, Zhang J, Guo G. Electromagnetic pulse exposure induces neuroinflammation and blood-brain barrier disruption by activating the NLRP3 inflammasome/NF-κB signaling pathway in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117972. [PMID: 40020384 DOI: 10.1016/j.ecoenv.2025.117972] [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: 11/27/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
The electromagnetic pulse (EMP) is a widespread electromagnetic disturbance that can disrupt electronic systems and pose health risks to personnel in operational areas. The biological effects of EMP radiation, especially on the central nervous system (CNS), are not yet fully understood but are gaining attention. This study examines the impact of EMP on the CNS using microglial cells as a model system. We found that mice exposed to a field strength of 600 kV/m with 1000 pulses per day for two weeks exhibited increased levels of oxidative stress. This exposure induced a microglia polarization to the M1 state, leading to neuroinflammation and disruption of the blood-brain barrier (BBB) by the pro-inflammatory response of microglia. Further analysis revealed that the NLRP3 inflammasome/NF-κB signaling pathway modulates the pro-inflammatory mechanisms of EMP irradiation. In conclusion, our findings show that EMP irradiation triggers neuroinflammation and BBB damage via NLRP3 inflammasome/NF-κB activation. This research highlights the effects of EMP radiation on the CNS and offers valuable insights into the potential targets for biomedical protection against it.
Collapse
Affiliation(s)
- Yanyun Lin
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Haiyang Lang
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China; The Ministry-of-Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Airforce Medical University, Xi'an 710032, China
| | - Peng Gao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Xia Miao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Qiyan Guo
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Yue Hao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Tao Ai
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Jing Li
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Jie Zhang
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China.
| | - Guozhen Guo
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China.
| |
Collapse
|
20
|
Ding X, Chen C, Zhao H, Dai B, Ye L, Song T, Huang S, Wang J, You T. Inhibiting SHP2 reduces glycolysis, promotes microglial M1 polarization, and alleviates secondary inflammation following spinal cord injury in a mouse model. Neural Regen Res 2025; 20:858-872. [PMID: 38886958 PMCID: PMC11433905 DOI: 10.4103/nrr.nrr-d-23-01925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/02/2024] [Accepted: 04/17/2024] [Indexed: 06/20/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202503000-00030/figure1/v/2024-06-17T092413Z/r/image-tiff Reducing the secondary inflammatory response, which is partly mediated by microglia, is a key focus in the treatment of spinal cord injury. Src homology 2-containing protein tyrosine phosphatase 2 (SHP2), encoded by PTPN11, is widely expressed in the human body and plays a role in inflammation through various mechanisms. Therefore, SHP2 is considered a potential target for the treatment of inflammation-related diseases. However, its role in secondary inflammation after spinal cord injury remains unclear. In this study, SHP2 was found to be abundantly expressed in microglia at the site of spinal cord injury. Inhibition of SHP2 expression using siRNA and SHP2 inhibitors attenuated the microglial inflammatory response in an in vitro lipopolysaccharide-induced model of inflammation. Notably, after treatment with SHP2 inhibitors, mice with spinal cord injury exhibited significantly improved hind limb locomotor function and reduced residual urine volume in the bladder. Subsequent in vitro experiments showed that, in microglia stimulated with lipopolysaccharide, inhibiting SHP2 expression promoted M2 polarization and inhibited M1 polarization. Finally, a co-culture experiment was conducted to assess the effect of microglia treated with SHP2 inhibitors on neuronal cells. The results demonstrated that inflammatory factors produced by microglia promoted neuronal apoptosis, while inhibiting SHP2 expression mitigated these effects. Collectively, our findings suggest that SHP2 enhances secondary inflammation and neuronal damage subsequent to spinal cord injury by modulating microglial phenotype. Therefore, inhibiting SHP2 alleviates the inflammatory response in mice with spinal cord injury and promotes functional recovery postinjury.
Collapse
Affiliation(s)
- Xintian Ding
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui Province, China
| | - Chun Chen
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Heng Zhao
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Bin Dai
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Lei Ye
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
- Department of Orthopedics, Provincial Hospital Affiliated to Anhui Medical University, Hefei, Anhui Province, China
| | - Tao Song
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Shuai Huang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Jia Wang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Tao You
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
21
|
Yu L, Sun L, Yu T, Guo A, Wu J, Chen J, Wang Q. CPCGI Alleviates Neural Damage by Modulating Microglial Pyroptosis After Traumatic Brain Injury. CNS Neurosci Ther 2025; 31:e70322. [PMID: 40059065 PMCID: PMC11890976 DOI: 10.1111/cns.70322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major global cause of mortality and long-term disability, with limited therapeutic options. Microglial pyroptosis, a form of programmed cell death associated with inflammation, has been implicated in exacerbating neuroinflammation and secondary injury following TBI. Compound porcine cerebroside ganglioside injection (CPCGI) has shown anti-inflammatory and antioxidant properties, but its effects on pyroptosis remain unexplored. This study investigates the role of CPCGI in TBI and its underlying mechanisms. METHODS A controlled cortical impact (CCI) model was utilized to establish TBI in vivo, while lipopolysaccharide (LPS) was used in vitro to induce microglial activation that mimicked TBI conditions. The effects of CPCGI on microglial pyroptosis and inflammatory cytokines were analyzed through immunofluorescence, flow cytometry, western blotting, and quantitative real-time PCR (qRT-PCR). The involvement of the NLRP3 inflammasome in CPCGI's mechanism was examined using NLRP3 overexpression or the NLRP3 agonist BMS-986299. A microglia-neuron interaction model was created, and neuronal injury was assessed with the Cell Counting Kit-8 and Fluoro-Jade C (FJC). RESULTS Treatment with CPCGI resulted in significant improvement in the neurobehavioral outcomes, reduced lesion volume, and decreased neuronal loss following TBI. Notably, TBI induced microglial pyroptosis and the release of pro-inflammatory cytokines, while CPCGI inhibited microglial pyroptosis, thereby mitigating the inflammatory response and reducing neuronal damage. Mechanistically, overexpression of NLRP3 in microglial cells reversed the inhibitory effects of CPCGI on microglial pyroptosis, indicating that CPCGI's inhibition of microglial pyroptosis may be mediated by the NLRP3 inflammasome. Furthermore, NLRP3 overexpression or administration of the NLRP3 agonist BMS-986299 negated the neuroprotective effects of CPCGI in vivo and in vitro. CONCLUSION These findings suggest that CPCGI provides neuroprotection in TBI by targeting NLRP3 inflammasome-mediated microglial pyroptosis, thereby improving the neuroinflammatory microenvironment and promoting neurological recovery. This underscores its potential as a promising candidate for TBI treatment.
Collapse
Affiliation(s)
- Lu‐Lu Yu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Lei Sun
- Department of NeurologyZhengzhou University People's HospitalZhengzhouChina
| | - Ting‐Ting Yu
- Department of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - An‐Chen Guo
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
| | - Jian‐Ping Wu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Jun‐Min Chen
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qun Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
22
|
John RK, Vogel SP, Zia S, Lee KV, Nguyen AT, Torres-Espin A, Fenrich KK, Ng C, Schmidt EKA, Vavrek R, Raposo PJF, Smith K, Fouad K, Plemel JR. Reawakening inflammation in the chronically injured spinal cord using lipopolysaccharide induces diverse microglial states. J Neuroinflammation 2025; 22:56. [PMID: 40022205 PMCID: PMC11871772 DOI: 10.1186/s12974-025-03379-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Rehabilitative training is an effective method to promote recovery following spinal cord injury (SCI), with lower training efficacy observed in the chronic stage. The increased training efficacy during the subacute period is associated with a shift towards a more adaptive or proreparative state induced by the SCI. A potential link is SCI-induced inflammation, which is elevated in the subacute period, and, as injection of lipopolysaccharide (LPS) alongside training improves recovery in chronic SCI, suggesting LPS could reopen a window of plasticity late after injury. Microglia may play a role in LPS-mediated plasticity as they react to LPS and are implicated in facilitating recovery following SCI. However, it is unknown how microglia change in response to LPS following SCI to promote neuroplasticity. MAIN BODY Here we used single-cell RNA sequencing to examine microglial responses in subacute and chronic SCI with and without an LPS injection. We show that subacute SCI is characterized by a disease-associated microglial (DAM) signature, while chronic SCI is highly heterogeneous, with both injury-induced and homeostatic states. DAM states exhibit predicted metabolic pathway activity and neuronal interactions that are associated with potential mediators of plasticity. With LPS injection, microglia shifted away from the homeostatic signature to a primed, translation-associated state and increased DAM in degenerated tracts caudal to the injury. CONCLUSION Microglial states following an inflammatory stimulus in chronic injury incompletely recapitulate the subacute injury environment, showing both overlapping and distinct microglial signatures across time and with LPS injection. Our results contribute to an understanding of how microglia and LPS-induced neuroinflammation contribute to plasticity following SCI.
Collapse
Affiliation(s)
- Rebecca K John
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sadie P Vogel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Kelly V Lee
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Antoinette T Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Carmen Ng
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Emma K A Schmidt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Pamela J F Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Keira Smith
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| |
Collapse
|
23
|
Zhu X, Wang X, Su H, Shi X, Zhang Y, Chai Y, Yuan R. Spatial Confinement-Enhanced Electrochemiluminescence of Gold Nanoclusters on 3D Porous ZrO 2 Hollow Nanospheres for the Assessment of Acute Myocardial Infarction Protein Markers. Anal Chem 2025; 97:3671-3679. [PMID: 39921617 DOI: 10.1021/acs.analchem.4c06278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025]
Abstract
Herein, the gold nanoclusters@three-dimensional (3D) porous ZrO2 hollow nanospheres (Au NCs@ZrO2) with spatial confinement-enhanced electrochemiluminescence (SCE-ECL) were first prepared to fabricate a biosensing platform for the ultrasensitive detection of insulin-like growth factor 1 (IGF-1), which was associated with cardiovascular disease, malignant tumor, and neuropathic pain. Specifically, the confinement of Au NCs in a 3D microenvironment significantly boosted the optical radiation of excited Au NCs because the vibration of ligand molecules was restricted, and the recombination of holes and electrons of excited Au NCs was facilitated in the optical process for enhancing ECL efficiency, resulting in 5.1-fold stronger ECL efficiency than Au NCs. As a proof of concept, based on Au NCs@ZrO2 as an emitter and an orderly and localized catalytic hairpin self-assembly (OL-CHA) system as a signal amplifier, the built ECL biosensing platform achieved fast and trace determination of IGF-1 with the detection limit (LOD) down to 0.36 fg/mL. Moreover, the ECL platform realized the assessment of the IGF-1 expression of acute myocardial infarction (AMI) patients and exhibited a more prominent accuracy than the enzyme-linked immunosorbent assay (ELISA). This work proposed a neoteric avenue for developing highly efficient ECL emitters, which presented a promising prospect in ultrasensitive bioanalysis for early diagnosis of diseases.
Collapse
Affiliation(s)
- Xiaochun Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| | - Xiaofeng Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| | - Huimei Su
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| | - Xiaoyu Shi
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| | - Yue Zhang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, P. R. China
| | - Yaqin Chai
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, P. R China
| |
Collapse
|
24
|
Li H, Zhang P, Lin M, Li K, Zhang C, He X, Gao K. Pyroptosis: candidate key targets for mesenchymal stem cell-derived exosomes for the treatment of bone-related diseases. Stem Cell Res Ther 2025; 16:68. [PMID: 39940049 PMCID: PMC11816542 DOI: 10.1186/s13287-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Bone-related diseases impact a large portion of the global population and, due to their high disability rates and limited treatment options, pose significant medical and economic challenges. Mesenchymal stem cells (MSCs) can differentiate into multiple cell types and offer strong regenerative potential, making them promising for treating various diseases. However, issues with the immune response and cell survival limit the effectiveness of cell transplantation. This has led to increased interest in cell-free stem cell therapy, particularly the use of exosomes, which is the most studied form of this approach. Exosomes are extracellular vesicles that contain proteins, lipids, and nucleic acids and play a key role in cell communication and material exchange. Pyroptosis, a form of cell death involved in innate immunity, is also associated with many diseases. Studies have shown that MSC-derived exosomes have therapeutic potential for treating a range of conditions by regulating inflammation and pyroptosis. This study explored the role of MSC-derived exosomes in modulating pyroptosis to improve the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Haiming Li
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Peng Zhang
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Minghui Lin
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Kang Li
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Cunxin Zhang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Xiao He
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Kai Gao
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China.
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| |
Collapse
|
25
|
Zha X, Zheng G, Skutella T, Kiening K, Unterberg A, Younsi A. Microglia: a promising therapeutic target in spinal cord injury. Neural Regen Res 2025; 20:454-463. [PMID: 38819048 PMCID: PMC11317945 DOI: 10.4103/nrr.nrr-d-23-02044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/23/2024] [Accepted: 03/22/2024] [Indexed: 06/01/2024] Open
Abstract
Microglia are present throughout the central nervous system and are vital in neural repair, nutrition, phagocytosis, immunological regulation, and maintaining neuronal function. In a healthy spinal cord, microglia are accountable for immune surveillance, however, when a spinal cord injury occurs, the microenvironment drastically changes, leading to glial scars and failed axonal regeneration. In this context, microglia vary their gene and protein expression during activation, and proliferation in reaction to the injury, influencing injury responses both favorably and unfavorably. A dynamic and multifaceted injury response is mediated by microglia, which interact directly with neurons, astrocytes, oligodendrocytes, and neural stem/progenitor cells. Despite a clear understanding of their essential nature and origin, the mechanisms of action and new functions of microglia in spinal cord injury require extensive research. This review summarizes current studies on microglial genesis, physiological function, and pathological state, highlights their crucial roles in spinal cord injury, and proposes microglia as a therapeutic target.
Collapse
Affiliation(s)
- Xiaowei Zha
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
26
|
Varsamos I, Patilas C, Galanis A, Zachariou D, Tsalimas G, Sakellariou E, Spyrou I, Rozis M, Kaspiris A, Karampinas PK, Vasiliadis E, Pneumaticos SG. The Impact of Nuclear Factor Kappa B on the Response of Microglia in Spinal Cord Injuries. Cureus 2025; 17:e79367. [PMID: 40125122 PMCID: PMC11929549 DOI: 10.7759/cureus.79367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Spinal cord injury (SCI) results in both primary and secondary damage, each contributing to the overall injury and its consequences. Following SCI, microglia, the resident immune cells of the central nervous system (CNS), undergo a series of complex responses that contribute to the pathophysiology of the injury. In the context of SCI, nuclear factor kappa B (NF-kB) emerged as a critical mediator in the regulation of inflammatory responses following SCI. The aim of this review is to provide a comprehensive understanding of the involvement of NF-kB in the response of microglia following SCI. The PUBMED database was searched using the following keywords: NF-kB AND microglia AND spinal cord injury. Clinical and experimental studies evaluating the role of NF-kB in the response of microglia following SCI were included. Systematic reviews, case reports, research protocols, conference articles, and studies in languages other than English were excluded. The final analysis included 52 studies. NF-kB signaling exerts profound effects on the microglial response following SCI, influencing the inflammatory milieu, tissue damage, and potential for repair and recovery. Deactivation of the NF-kB signaling pathway suppresses the production of proinflammatory mediators in microglia, after SCI. Moreover, NF-kB suppression has neuroprotective effects, as it mitigates neuronal apoptosis and facilitates the M2 microglial phenotype, alleviating tissue damage after SCI. Moreover, several microRNAs play a crucial role in regulating gene expression post-transcriptionally and have emerged as key regulators in microglia activation after SCI. Overall, the role of NF-kB in the response of microglia to SCI is complex and context-dependent. While NF-kB activation is involved in initiating and propagating the inflammatory response following SCI, it also plays a role in tissue repair and regeneration. Thus, modulating NF-kB signaling in microglia represents a potential therapeutic target for attenuating inflammation and promoting neuroprotection and tissue repair in SCI.
Collapse
Affiliation(s)
- Iordanis Varsamos
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Christos Patilas
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Athanasios Galanis
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Dimitrios Zachariou
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Georgios Tsalimas
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Evangelos Sakellariou
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Ioannis Spyrou
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Meletis Rozis
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Angelos Kaspiris
- Division for Orthopaedic Research, Laboratory of Molecular Pharmacology, School of Health Sciences, University of Patras, Patras 26504, Greece, Patras, GRC
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Panayiotis K Karampinas
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Elias Vasiliadis
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| | - Spyros G Pneumaticos
- 3rd Department of Orthopedics, National and Kapodistrian University of Athens, KAT General Hospital, Athens, GRC
| |
Collapse
|
27
|
Quan L, Uyeda A, Manabe I, Muramatsu R. Astrocytic heterogeneous nuclear ribonucleoprotein U is involved in scar formation after spinal cord injury. J Neuroinflammation 2025; 22:28. [PMID: 39891176 PMCID: PMC11786496 DOI: 10.1186/s12974-025-03351-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025] Open
Abstract
Astrocytes have a beneficial role in tissue repair after central nervous system (CNS) injury. Although astrocyte proliferation is activated in response to injury, the intracellular mechanisms of astrocyte proliferation during acute phase of injury are not fully clarified. In this study, by functionally screening the highly expressed genes in the pathological state of spinal astrocytes, heterogeneous nuclear ribonucleoprotein U (Hnrnpu) is identified as a potential endogenous molecule that regulates astrocyte proliferation and the following scar formation. Inhibition of Hnrnpu in astrocytes impairs the formation of astrocytic glial scar, motor function recovery, and neuronal regeneration after spinal cord injury (SCI) in mice. In human astrocytes, HNRNPU knockdown downregulates the genes related to the astrocyte functions in scar formation and neuronal regeneration. These findings uncover that modulation of endogenous astrocytic function would be a promising therapeutic avenue to restore neurological function after CNS injury.
Collapse
Affiliation(s)
- Lili Quan
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Ichiro Manabe
- Department of Systems Medicine, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan.
| |
Collapse
|
28
|
Dong Q, Chen P, Qiu W, Yang Z, Li Y, Zhou Y, Guo L, Zou D, Xu R, Gao M. Long non-coding RNA Malat1 modulates CXCR4 expression to regulate the interaction between induced neural stem cells and microglia following closed head injury. Stem Cell Res Ther 2025; 16:31. [PMID: 39881403 PMCID: PMC11780782 DOI: 10.1186/s13287-024-04116-1] [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: 07/31/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Closed head injury (CHI) provokes a prominent neuroinflammation that may lead to long-term health consequences. Microglia plays pivotal and complex roles in neuroinflammation-mediated neuronal insult and repair following CHI. We previously reported that induced neural stem cells (iNSCs) can block the effects of CXCL12/CXCR4 signaling on NF-κB activation in activated microglia by CXCR4 overexpression. Here we aim to uncover the mechanism of CXCR4 upregulation in iNSCs. METHODS We performed bioinformatic analysis to detect the differentially expressed genes in iNSCs after co-cultured with LPS-activated microglia. Subsequently, we predicted the target genes and performed gain- and loss-of-functional studies, dualluciferase reporter, RNA immunoprecipitation, biotin-coupled miRNA pulldown, fluorescence in situ hybridization and cell transplantation assays to further elucidate the mechanism underlying the immunoregulatory effects of iNSCs. Student's t-test and one-way analysis of variance (ANOVA) with Tukey's post hoc test were used to determine statistical significance. RESULTS Our results indicated that Malat1 could act as a sponge of miR-139-5p to modulate the expression of CXCR4 that exerted significant influence on the immunoregulatory effects of iNSCs on the secretion of CXCL12, TNF-α and IGF-1 by activated microglia. Furthermore, Malat1 inhibition blocked the immunoregulatory effects of iNSC grafts on microglial activation as well as neuroinflammation in the injured cortices of CHI mice. Interestingly, NF-κB activation in iNSCs augmented the immunoregulatory effects of iNSCs on microglial activation by activating the axis of Malat1/miR-139-5p/Cxcr4. Notably, we found that TNF-α secreted by activated microglia could bind to TNFR1 at the surface of iNSCs to trigger NF-κB activation in iNSCs. CONCLUSIONS In short, our findings reveal a novel role of Malat1 in the immunomodulatory effects of iNSCs on microglial activation, suggesting that transplanted iNSCs may self-perceive the changes of the activated state of microglia and thus make prudential regulation of the neuroinflammation following CHI.
Collapse
Affiliation(s)
- Qin Dong
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, 100038, China
| | - Pengyu Chen
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province, 350122, China
| | - Wenqiao Qiu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| | - Zhijun Yang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
- Hebei Leren Biotechnology Co., LTD, Shijiazhuang, Hebei Province, 050000, China
| | - Yanteng Li
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yuhui Zhou
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
- Chuanxing Middle School, Xichang, Sichuan Province, 615000, China
| | - Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| | - Dan Zou
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, School of Medicine, Sichuan Academy of Medical Sciences, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, 610072, China.
| | - Mou Gao
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
29
|
Timofeeva AV, Akhmetzyanova ER, Rizvanov AA, Mukhamedshina YO. Interaction of microglia with the microenvironment in spinal cord injury. Neuroscience 2025; 565:594-603. [PMID: 39622381 DOI: 10.1016/j.neuroscience.2024.11.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
This article discusses the peculiarities of microglia behaviour and their interaction with other cells of the central nervous system (CNS) during neural tissue injury with a focus on spinal cord injury (SCI). Taking into account the plasticity of microglia, the influence of the microenvironment should be taken into account to establish the mechanisms determining the polarization pathways of these cells. Determination of the system of microglia interactions with other CNS cells during injury will reveal the patterns of post-traumatic microglia responses, in particular, determining both pro-inflammatory and anti-inflammatory responses. This review compiles information on changes in microglia activation, migration and phagocytosis, as well as their reciprocal effects on other CNS cells, such as neurons, astrocytes and oligodendrocytes, in the background of SCI. The information contained in this article may be of interest not only to scientists studying traumatic injuries of the central nervous system, but also to specialists in the field of studying and treating neurodegenerative diseases, since the mechanisms occurring in the injured spinal cord may also be characteristic of pathological events in degenerative processes.
Collapse
Affiliation(s)
- A V Timofeeva
- Kazan (Volga Region) Federal University, Kazan, Russia
| | | | - A A Rizvanov
- Kazan (Volga Region) Federal University, Kazan, Russia; Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Russia
| | - Y O Mukhamedshina
- Kazan (Volga Region) Federal University, Kazan, Russia; Kazan State Medical University, Kazan, Russia
| |
Collapse
|
30
|
Ouyang F, Zheng M, Li J, Huang J, Ye J, Wang J, Zhao Y, Shan F, Li Z, Yu S, Yao F, Tian D, Cheng L, Jing J. Glycosylated lysosomal membrane protein promotes tissue repair after spinal cord injury by reducing iron deposition and ferroptosis in microglia. Sci Rep 2025; 15:2867. [PMID: 39843796 PMCID: PMC11754889 DOI: 10.1038/s41598-025-86991-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Excessive iron deposition can lead to ferroptosis, a form of iron-dependent cell death detrimental to neuronal survival. Microglia have been identified as having a high capacity for iron deposition, yet it remains unclear whether microglia undergo ferroptosis while phagocytosing excessive amounts of iron after spinal cord injury (SCI). Here, we observed scattered iron around the epicenter of the injured spinal cord at 7 days post-injury (dpi) in mice, which then accumulated in the lesion core at 14 dpi. Concurrently, microglia exhibited elevated expression of the iron-storage protein ferritin and were found to undergo ferroptosis between 7 and 28 dpi. Additionally, we noted a gradual decrease in glycosylated lysosomal membrane protein (GLMP) which is associated with iron metabolism in microglia undergoing ferroptosis. In situ injection of AAV9-Cx3cr1-shGlmp-eGFP to knock down GLMP specifically in microglia resulted in a significant increase in iron deposition and ferroptosis, leading to an expanded lesion area, aggravated neuronal loss, and subsequent inhibition of functional restoration. Our findings highlight the crucial role of GLMP in mitigating iron overload and ferroptosis in microglia, thereby contributing to axon retention and locomotor recovery after SCI.
Collapse
Affiliation(s)
- Fangru Ouyang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jianjian Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jinxin Huang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianan Ye
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jingwen Wang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuanzhe Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fangli Shan
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ziyu Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Shuishen Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Dasheng Tian
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- Institute of Orthopedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
31
|
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
|
32
|
Zhang H, Xiang L, Yuan H, Yu H. ARL11 knockdown alleviates spinal cord injury by inhibiting neuroinflammation and M1 activation of microglia in mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167522. [PMID: 39307293 DOI: 10.1016/j.bbadis.2024.167522] [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: 04/10/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024]
Abstract
Spinal cord injury (SCI) is a severe central nervous system injury and microglia are major participants in neuroinflammation after injury. ADP-ribosylation factor-like GTPase 11 (ARL11) is a GTP-binding protein. Whether ARL11 is involved in the SCI progression is unknown. In the impactor-induced moderate SCI mouse model, ARL11 protein and mRNA expression were significantly increased in the injury site. LPS (100 ng/mL) and IFN-γ (20 ng/mL) were incubated with BV2 cells (immortalized mouse microglial cell line) to drive them into an M1-like phenotype. ARL11 up-regulation was also observed in activated microglia in SCI mice and LPS and IFN-γ treated BV2 cells. Basso Mouse Scale scores and inclined plate test revealed that ARL11 deletion promoted motor function recovery in SCI mice. Pathological examination showed ARL11 knockdown reduced spinal cord tissue damage, increased neuron numbers, and inhibited neuronal apoptosis in SCI mice. ARL11 knockdown notably inhibited IL-1β and IL-6 production in vivo and in vitro. Furthermore, ARL11 deletion significantly inhibited iNOS protein and mRNA expression in vivo and in vitro, and COX-2 expression in vivo. Mechanism studies revealed that ARL11 silencing decreased phosphorylated ERK1/2 protein expression. Additionally, ELF1 knockdown significantly inhibited ARL11 protein and mRNA expression in vitro. ELF1 acted as a transcription activator in regulating ARL11 expression by binding to the promoter. In conclusion, ARL11 knockdown protects neurons by inhibiting M1 microglia-induced neuroinflammation, thereby promoting motor functional recovery in SCI mice. This may occur in part under the regulation of ELF1. Our study provides a new molecular target for SCI treatment.
Collapse
Affiliation(s)
- Haocong Zhang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China
| | - Liangbi Xiang
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China
| | - Hong Yuan
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China
| | - Hailong Yu
- Department of Orthopaedics, The General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, Liaoning, China.
| |
Collapse
|
33
|
Stewart AN, Bosse-Joseph CC, Kumari R, Bailey WM, Park KA, Slone VK, Gensel JC. Nonresolving Neuroinflammation Regulates Axon Regeneration in Chronic Spinal Cord Injury. J Neurosci 2025; 45:e1017242024. [PMID: 39510834 DOI: 10.1523/jneurosci.1017-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/17/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic spinal cord injury (SCI) lesions retain increased densities of microglia and macrophages. In acute SCI, macrophages induce growth cone collapse and facilitate axon retraction away from lesion boundaries. Little is known about the role of sustained inflammation in chronic SCI or whether chronic inflammation affects regeneration. We used the colony-stimulating factor-1 receptor inhibitor, PLX-5622, to deplete microglia and macrophages months after complete crush SCI in female mice. Transcriptional analyses revealed a significant inflammatory depletion within chronic SCI lesions after PLX-5622 treatment. Both transcriptional analyses and immunohistochemistry revealed that Iba1+ cells repopulate to predepleted densities after treatment removal. Neuronal-enriched transcripts were significantly elevated in mice after inflammatory repopulation, but no significant effects were observed with inflammatory depletion alone. Axon densities also significantly increased within the lesion after PLX-5622 treatment and after repopulation. To better examine the effect of chronic inflammation on axon regeneration, we tested PLX-5622 treatment in neuronal-specific phosphatase and tensin homolog protein (PTEN) knock-out (PTEN-KO) mice. PTEN-KO was delivered using spinal injections of retrogradely transported adeno-associated viruses (AAVrg's). PTEN-KO did not further increase axon densities within the lesion beyond the effects induced by PLX-5622. Axons that grew within the lesion were histologically identified as 5-HT+ and CGRP+, both of which are not robustly transduced by AAVrg's. Our work identified that increased macrophage/microglial densities in the chronic SCI environment may be actively retained by homeostatic mechanisms likely affiliated with a sustained elevated expression of CSF1 and other chemokines. Finally, we identify a novel role of sustained inflammation as a prospective barrier to axon regeneration in chronic SCI.
Collapse
Affiliation(s)
- Andrew N Stewart
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Christopher C Bosse-Joseph
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Reena Kumari
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | - William M Bailey
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| | - Kennedy A Park
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - Victoria K Slone
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky 40536
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
34
|
Liu Z, Xiang C, Zhao X, Aizawa T, Niu R, Zhao J, Guo F, Li Y, Luo W, Liu W, Gu R. Regulation of dynamic spatiotemporal inflammation by nanomaterials in spinal cord injury. J Nanobiotechnology 2024; 22:767. [PMID: 39696584 DOI: 10.1186/s12951-024-03037-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/24/2024] [Indexed: 12/20/2024] Open
Abstract
Spinal cord injury (SCI) is a common clinical condition of the central nervous system that can lead to sensory and motor impairment below the injury level or permanent loss of function in severe cases. Dynamic spatiotemporal neuroinflammation is vital to neurological recovery, which is collectively constituted by the dynamic changes in a series of inflammatory cells, including microglia, neutrophils, and astrocytes, among others. Immunomodulatory nanomaterials can readily improve the therapeutic effects and simultaneously overcome various drawbacks associated with treatment, such as the off-target side effects and loss of bioactivity of immune agents during circulation. In this review, we discuss the role of dynamic spatiotemporal inflammation in secondary injuries after SCI, elaborate on the mechanism of action and effect of existing nanomaterials in treating SCI, and summarize the mechanism(s) whereby they regulate inflammation. Finally, the challenges and prospects associated with using nanotechnology to modulate immunotherapy are discussed to provide new insights for future treatment. Deciphering the intricate spatiotemporal mechanisms of neuroinflammation in SCI requires further in-depth studies. Therefore, SCI continues to represent a formidable challenge.
Collapse
Affiliation(s)
- Zeping Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Chunyu Xiang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Xu Zhao
- Department of Orthopedics, Third Military Medical University, Xinqiao Hosp, 83 Xinqiao Main St, Chongqing, 400037, PR China
| | - Toshimi Aizawa
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Renrui Niu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Jianhui Zhao
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Fengshuo Guo
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Yueying Li
- Department of Hand & Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Wenqi Luo
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| | - Wanguo Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| | - Rui Gu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| |
Collapse
|
35
|
Dhir S, Derue H, Ribeiro-da-Silva A. Temporal changes of spinal microglia in murine models of neuropathic pain: a scoping review. Front Immunol 2024; 15:1460072. [PMID: 39735541 PMCID: PMC11671780 DOI: 10.3389/fimmu.2024.1460072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/18/2024] [Indexed: 12/31/2024] Open
Abstract
Neuropathic pain (NP) is an ineffectively treated, debilitating chronic pain disorder that is associated with maladaptive changes in the central nervous system, particularly in the spinal cord. Murine models of NP looking at the mechanisms underlying these changes suggest an important role of microglia, the resident immune cells of the central nervous system, in various stages of disease progression. However, given the number of different NP models and the resource limitations that come with tracking longitudinal changes in NP animals, many studies fail to truly recapitulate the patterns that exist between pain conditions and temporal microglial changes. This review integrates how NP studies are being carried out in murine models and how microglia changes over time can affect pain behavior in order to inform better study design and highlight knowledge gaps in the field. 258 peer-reviewed, primary source articles looking at spinal microglia in murine models of NP were selected using Covidence. Trends in the type of mice, statistical tests, pain models, interventions, microglial markers and temporal pain behavior and microglia changes were recorded and analyzed. Studies were primarily conducted in inbred, young adult, male mice having peripheral nerve injury which highlights the lack of generalizability in the data currently being collected. Changes in microglia and pain behavior, which were both increased, were tested most commonly up to 2 weeks after pain initiation despite aberrant microglia activity also being recorded at later time points in NP conditions. Studies using treatments that decrease microglia show decreased pain behavior primarily at the 1- and 2-week time point with many studies not recording pain behavior despite the involvement of spinal microglia dysfunction in their development. These results show the need for not only studying spinal microglia dynamics in a variety of NP conditions at longer time points but also for better clinically relevant study design considerations.
Collapse
Affiliation(s)
- Simran Dhir
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Hannah Derue
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
36
|
Shelton DA, Papania JT, Getz TE, Sellers JT, Giradot PE, Chrenek MA, Grossniklaus HE, Boatright JH, Nickerson JM. Loss of Pigment Epithelium Derived Factor Sensitizes C57BL/6J Mice to Light-Induced Retinal Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626802. [PMID: 39679905 PMCID: PMC11643041 DOI: 10.1101/2024.12.04.626802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Purpose Pigment epithelium-derived factor (PEDF) is a neurotrophic glycoprotein secreted by the retinal pigment epithelium (RPE) that supports retinal photoreceptor health. Deficits in PEDF are associated with increased inflammation and retinal degeneration in aging and diabetic retinopathy. We hypothesized that light-induced stress in C57BL/6J mice deficient in PEDF would lead to increased retinal neuronal and RPE defects, impaired expression of neurotrophic factor Insulin-like growth factor 1 (IGF-1), and overactivation of Galectin-3-mediated inflammatory signaling. Methods C57BL/6J mice expressing the RPE65 M450/M450 allele were crossed to PEDF KO/KO and wildtype (PEDF +/+) littermates. Mice were exposed to 50,000 lux light for 5 hours to initiate acute damage. Changes in visual function outcomes were tracked via electroretinogram (ERG), confocal scanning laser ophthalmoscopy(cSLO), and spectral domain optical coherence tomography (SD-OCT) on days 3, 5, and 7 post-light exposure. Gene and protein expression of Galectin-3 were measured by digital drop PCR (ddPCR) and western blot. To further investigate the role of galectin-3 on visual outcomes and PEDF expression after damage, we also used a small-molecule inhibitor to reduce its activity. Results Following light damage, PEDF KO/KO mice showed more severe retinal thinning, impaired visual function (reduced a-, b-, and c-wave amplitudes), and increased Galectin-3 expressing immune cell infiltration compared to PEDF +/+. PEDF KO/KO mice had suppressed damage-associated increases in IGF-1 expression. Additionally, baseline Galectin-3 mRNA and protein expression were reduced in PEDF KO/KO mice compared to PEDF +/+. However, after light damage, Galectin-3 expression decreases in PEDF +/+ mice but increases in PEDF KO/KO mice without reaching PEDF +/+ levels. Galectin-3 inhibition worsens retinal degeneration, reduces PEDF expression in PEDF +/+ mice, and mimics the effects seen in PEDF knockouts. Conclusions Loss of PEDF alone does not elicit functional defects in C57BL/6J mice. However, under light stress, PEDF deficiency significantly increases severe retinal degeneration, visual deficits, Galectin-3 expression, and suppression of IGF-1 than PEDF +/+. PEDF deficiency reduced baseline expression of Galectin-3, and pharmacological inhibition of Galectin-3 worsens outcomes and suppresses PEDF expression in PEDF +/+, suggesting a novel co-regulatory relationship between the two proteins in mitigating light-induced retinal damage.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Tatiana E. Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jana T. Sellers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Preston E. Giradot
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | | | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
37
|
Cao L, Huang X, Zhu J, Xiao J, Xie L. Falcarindiol improves functional recovery and alleviates neuroinflammation after spinal cord injury by inhibiting STAT/MAPK signaling pathways. Biochem Biophys Res Commun 2024; 736:150860. [PMID: 39454306 DOI: 10.1016/j.bbrc.2024.150860] [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: 06/16/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Spinal cord injury (SCI) is a devastating trauma in the central nervous system (CNS), leading to motor and sensory impairment. Neuroinflammation is one of the critical contributors to the progression of secondary injury. Falcarindiol has been reported to efficaciously mitigate lipopolysaccharide (LPS)-mediated inflammation in RAW 264.7 cells. The role of falcarindiol in SCI recovery remains unclear. In this present study, traumatic SCI mice models and LPS-stimulated murine microglia cell line (BV2 cells) were performed to explore the pharmacological effects and the underlying mechanisms of falcarindiol in improving SCI repair with detection of motor function recovery, morphological changes, numbers of survival neurons and protein expression levels of inflammation or apoptosis-related proteins. Our study found that falcarindiol intervention could promote motor function recovery and reduce spinal cord tissue damage in mice following SCI. Mechanistically, falcarindiol intervention suppressed apoptosis-driven neuronal cell death and mitigated inflammatory reactions following SCI. Additionally, falcarindiol inhibited the activation of signal transducer and activator of transcription (STAT) and mitogen-activated protein kinases (MAPK) signaling pathways in vivo and in vitro. This suppression of STAT and MAPK activation by falcarindiol was reversed by STAT3 agonist Colivelin TFA and MAPK agonist C16-PAF in BV2 cells, respectively. Moreover, the study further demonstrated that the anti-inflammation role of falcarindiol was obstructed by Colivelin TFA but not by C16-PAF in LPS-stimulated BV2 cells, suggesting that falcarindiol may efficaciously ameliorate neuroinflammation through inhibiting the activation of STAT signaling pathway following SCI. Collectively, our study indicates that falcarindiol may be a novel drug candidate for the treatment and management of SCI.
Collapse
Affiliation(s)
- Lin Cao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Jinhua Maternal and Child Health Care Hospital, Jinhua, 321099, China
| | - Xiaoli Huang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiangwei Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Ling Xie
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
38
|
Dill-Macky AS, Lee EN, Wertheim JA, Koss KM. Glia in tissue engineering: From biomaterial tools to transplantation. Acta Biomater 2024; 190:24-49. [PMID: 39396630 DOI: 10.1016/j.actbio.2024.10.017] [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: 02/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Glia are imperative in nearly every function of the nervous system, including neurotransmission, neuronal repair, development, immunity, and myelination. Recently, the reparative roles of glia in the central and peripheral nervous systems have been elucidated, suggesting a tremendous potential for these cells as novel treatments to central nervous system disorders. Glial cells often behave as 'double-edged swords' in neuroinflammation, ultimately deciding the life or death of resident cells. Compared to glia, neuronal cells have limited mobility, lack the ability to divide and self-renew, and are generally more delicate. Glia have been candidates for therapeutic use in many successful grafting studies, which have been largely focused on restoring myelin with Schwann cells, olfactory ensheathing glia, and oligodendrocytes with support from astrocytes. However, few therapeutics of this class have succeeded past clinical trials. Several tools and materials are being developed to understand and re-engineer these grafting concepts for greater success, such as extra cellular matrix-based scaffolds, bioactive peptides, biomolecular delivery systems, biomolecular discovery for neuroinflammatory mediation, composite microstructures such as artificial channels for cell trafficking, and graft enhanced electrical stimulation. Furthermore, advances in stem cell-derived cortical/cerebral organoid differentiation protocols have allowed for the generation of patient-derived glia comparable to those acquired from tissues requiring highly invasive procedures or are otherwise inaccessible. However, research on bioengineered tools that manipulate glial cells is nowhere near as comprehensive as that for systems of neurons and neural stem cells. This article explores the therapeutic potential of glia in transplantation with an emphasis on novel bioengineered tools for enhancement of their reparative properties. STATEMENT OF SIGNIFICANCE: Neural glia are responsible for a host of developmental, homeostatic, and reparative roles in the central nervous system but are often a major cause of tissue damage and cellular loss in insults and degenerative pathologies. Most glial grafts have employed Schwann cells for remyelination, but other glial with novel biomaterials have been employed, emphasizing their diverse functionality. Promising strategies have emerged, including neuroimmune mediation of glial scar tissues and facilitated migration and differentiation of stem cells for neural replacement. Herein, a comprehensive review of biomaterial tools for glia in transplantation is presented, highlighting Schwann cells, astrocytes, olfactory ensheating glia, oligodendrocytes, microglia, and ependymal cells.
Collapse
Affiliation(s)
- A S Dill-Macky
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - E N Lee
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - J A Wertheim
- Department of Surgery, University of Arizona, 1501 N Campbell Ave, Tucson, AZ 85724, United States
| | - K M Koss
- Department of Neurobiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0625, United States; Sealy Institute for Drug Discovery, University of Texas Medical Branch, 105 11th Street Galveston, TX 77555-1110, United States.
| |
Collapse
|
39
|
Sámano C, Mazzone GL. The role of astrocytes response triggered by hyperglycaemia during spinal cord injury. Arch Physiol Biochem 2024; 130:724-741. [PMID: 37798949 DOI: 10.1080/13813455.2023.2264538] [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: 03/24/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE This manuscript aimed to provide a comprehensive overview of the physiological, molecular, and cellular mechanisms triggered by reactive astrocytes (RA) in the context of spinal cord injury (SCI), with a particular focus on cases involving hyperglycaemia. METHODS The compilation of articles related to astrocyte responses in neuropathological conditions, with a specific emphasis on those related to SCI and hyperglycaemia, was conducted by searching through databases including Science Direct, Web of Science, and PubMed. RESULTS AND CONCLUSIONS This article explores the dual role of astrocytes in both neurophysiological and neurodegenerative conditions within the central nervous system (CNS). In the aftermath of SCI and hyperglycaemia, astrocytes undergo a transformation into RA, adopting a distinct phenotype. While there are currently no approved therapies for SCI, various therapeutic strategies have been proposed to alleviate the detrimental effects of RAs following SCI and hyperglycemia. These strategies show promising potential in the treatment of SCI and its likely comorbidities.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa (UAM-C), Ciudad de México, México
| | - G L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| |
Collapse
|
40
|
Li J, Zheng M, Ouyang F, Ye J, Huang J, Zhao Y, Wang J, Shan F, Li Z, Yu S, Yao F, Tian D, Cheng L, Jing J. Interleukin-3 Modulates Macrophage Phagocytic Activity and Promotes Spinal Cord Injury Repair. CNS Neurosci Ther 2024; 30:e70181. [PMID: 39697159 DOI: 10.1111/cns.70181] [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: 06/02/2024] [Revised: 09/28/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Effective clearance of lipid-rich debris by macrophages is critical for neural repair and regeneration after spinal cord injury (SCI). Interleukin-3 (IL-3) has been implicated in programming microglia to cluster and clear pathological aggregates in neurodegenerative disease. Yet, the influence of IL-3 on lipid debris clearance post-SCI is not well characterized. METHODS We established a mouse model of spinal cord compression injury to investigate the role of IL-3. Blockage of IL-3 was achieved through intrathecal delivery of an IL-3-neutralizing antibody, while IL-3 activation was augmented via in situ injection of recombinant IL-3 into the lesion site immediately post-SCI. Immunofluorescence staining was performed to determine IL-3 and IL-3Rα sources and distribution, lipid droplet accumulation, neuron preservation, and axon regeneration after SCI. The Basso Mouse Scale (BMS) and footprint analysis were employed to evaluate locomotor function recovery. RESULTS We found that IL-3 expression was significantly upregulated post-SCI, peaking at 14 days post-injury (dpi) and persisting until 28 dpi. Notably, IL-3 was primarily secreted by astrocytes surrounding the lesion epicenter. Correspondingly, IL-3Rα was predominantly observed in macrophages within the injury core, also elevating at 14 dpi. Neutralization of IL-3 led to increased lipid droplet accumulation, along with markedly widespread of macrophages and decreased neuronal survival, resulting in severe motor deficits compared to controls. Conversely, in situ injection of IL-3 reduced lipid droplet accumulation in macrophages, preserved neurons, promoted axon regeneration, and ultimately contributed to the recovery of motor function after SCI. CONCLUSION Our findings shed light on the role of IL-3 in modulating macrophage phagocytic activity and suggest that the IL-3/IL-3Rα pathway may be a potential therapeutic target for enhancing neural repair and functional recovery after SCI.
Collapse
Affiliation(s)
- Jianjian Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meige Zheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangru Ouyang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianan Ye
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinxin Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanzhe Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingwen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangli Shan
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyu Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuishen Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
41
|
Latini L, De Araujo DSM, Amato R, Canovai A, Buccarello L, De Logu F, Novelli E, Vlasiuk A, Malerba F, Arisi I, Florio R, Asari H, Capsoni S, Strettoi E, Villetti G, Imbimbo BP, Dal Monte M, Nassini R, Geppetti P, Marinelli S, Cattaneo A. A p75 neurotrophin receptor-sparing nerve growth factor protects retinal ganglion cells from neurodegeneration by targeting microglia. Br J Pharmacol 2024; 181:4890-4919. [PMID: 39252503 DOI: 10.1111/bph.17316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Retinal ganglion cells (RGCs) are the output stage of retinal information processing, via their axons forming the optic nerve (ON). ON damage leads to axonal degeneration and death of RGCs, and results in vision impairment. Nerve growth factor (NGF) signalling is crucial for RGC operations and visual functions. Here, we investigate a new neuroprotective mechanism of a novel therapeutic candidate, a p75-less, TrkA-biased NGF agonist (hNGFp) in rat RGC degeneration, in comparison with wild type human NGF (hNGFwt). EXPERIMENTAL APPROACH Both neonate and adult rats, whether subjected or not to ON lesion, were treated with intravitreal injections or eye drops containing either hNGFp or hNGFwt. Different doses of the drugs were administered at days 1, 4 or 7 after injury for a maximum of 10 days, when immunofluorescence, electrophysiology, cellular morphology, cytokine array and behaviour studies were carried out. Pharmacokinetic evaluation was performed on rabbits treated with hNGFp ocular drops. RESULTS hNGFp exerted a potent RGC neuroprotection by acting on microglia cells, and outperformed hNGFwt in rescuing RGC degeneration and reducing inflammatory molecules. Delayed use of hNGFp after ON lesion resulted in better outcomes compared with treatment with hNGFwt. Moreover, hNGFp-based ocular drops were less algogenic than hNGFwt. Pharmacokinetic measurements revealed that biologically relevant quantities of hNGFp were found in the rabbit retina. CONCLUSIONS AND IMPLICATIONS Our data point to microglia as a new cell target through which NGF-induced TrkA signalling exerts neuroprotection of the RGC, emphasizing hNGFp as a powerful treatment to tackle retinal degeneration.
Collapse
Affiliation(s)
- Laura Latini
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Anastasiia Vlasiuk
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Hiroki Asari
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Simona Capsoni
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Gino Villetti
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
42
|
Tahmasian N, Feng MY, Arbabi K, Rusu B, Cao W, Kukreja B, Lubotzky A, Wainberg M, Tripathy SJ, Kalish BT. Neonatal Brain Injury Triggers Niche-Specific Changes to Cellular Biogeography. eNeuro 2024; 11:ENEURO.0224-24.2024. [PMID: 39681473 DOI: 10.1523/eneuro.0224-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Preterm infants are at risk for brain injury and neurodevelopmental impairment due, in part, to white matter injury following chronic hypoxia exposure. However, the precise molecular mechanisms by which neonatal hypoxia disrupts early neurodevelopment are poorly understood. Here, we constructed a brain-wide map of the regenerative response to newborn brain injury using high-resolution imaging-based spatial transcriptomics to analyze over 800,000 cells in a mouse model of chronic neonatal hypoxia. Additionally, we developed a new method for inferring condition-associated differences in cell type spatial proximity, enabling the identification of niche-specific changes in cellular architecture. We observed hypoxia-associated changes in region-specific cell states, cell type composition, and spatial organization. Importantly, our analysis revealed mechanisms underlying reparative neurogenesis and gliogenesis, while also nominating pathways that may impede circuit rewiring following neonatal hypoxia. Altogether, our work provides a comprehensive description of the molecular response to newborn brain injury.
Collapse
Affiliation(s)
- Nareh Tahmasian
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Min Yi Feng
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Bianca Rusu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Wuxinhao Cao
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Asael Lubotzky
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Michael Wainberg
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario M5G 1X5, Canada
| | - Shreejoy J Tripathy
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Brian T Kalish
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
43
|
Lin W, Cai Z, Liang J, Miao P, Ruan Y, Li P, Lin S, Tian H, Yu Q, He X. Heme Oxygenase-1 Overexpression Activates the IRF1/DRP1 Signaling Pathway to Promote M2-Type Polarization of Spinal Cord Microglia. Drug Dev Res 2024; 85:e70033. [PMID: 39660541 DOI: 10.1002/ddr.70033] [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: 07/16/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 12/12/2024]
Abstract
Microglia-mediated neuroinflammatory responses have a critical function in the spinal cord injury (SCI) mechanism, and targeted modulation of microglia activity has emerged as a new therapeutic strategy for SCI. Heme oxygenase 1(HO-1) regulates the close dynamic crosstalk between oxidative stress and inflammatory responses. This investigation aimed to study the molecular pathways by which HO-1 regulates the inflammatory response of microglia. We cultivated primary rat spinal cord microglia and BV2 cell lines and used lipopolysaccharide (LPS) to stimulate microglia to establish an in vitro model. The adeno-associated virus (AAV) was used to induce HO-1 overexpression to observe the effects of HO-1 overexpression on microglia survival, morphological changes, microglia activation, inflammatory cytokines secretion, mitochondrial dynamics, and nucleotide-binding oligomerization domain-like receptor protein (NLRP3) inflammatory complex and nuclear factor-κB (NF-κB) signaling pathways. It was found that HO-1 overexpression was successfully induced using an AAV on microglia in vitro. HO-1 overexpression increased microglia survival and reduced microglia apoptosis in the inflammatory microenvironment. Overexpressed HO-1 inhibited microglia M1-type polarization, downregulated the NF-κB signaling pathway, inhibited NLRP3 inflammatory complex activation, and reduced the secretion of inflammatory factors. Overexpressed HO-1 maintained the stability of mitochondrial dynamics and inhibited excessive mitochondrial cleavage. Further experiments showed that overexpression of HO-1 activated the interferon regulatory factor 1 (IRF1)/dynamin-related protein 1 (DRP1) signaling pathway, thereby promoting microglia M2-type polarization and improving neuronal survival. This study demonstrates that HO-1 activates the IRF1/DRP1 axis, promoting M2 polarization in microglia and attenuating neuroinflammation by suppressing the NF-κB signaling pathway. These outcomes offer new visions and important clues for effectively managing SCI in the clinic.
Collapse
Affiliation(s)
- Wenping Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ziming Cai
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jinzhu Liang
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Ping Miao
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ye Ruan
- Department of The Third School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pian Li
- Department of The Third School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhui Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - He Tian
- Department of Gynaecology, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qinghe Yu
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xu He
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
44
|
Ye Y, Su X, Tang J, Zhu C. Neuropathic Pain Induced by Spinal Cord Injury from the Glia Perspective and Its Treatment. Cell Mol Neurobiol 2024; 44:81. [PMID: 39607514 PMCID: PMC11604677 DOI: 10.1007/s10571-024-01517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Regional neuropathic pain syndromes above, at, or below the site of spinal damage arise after spinal cord injury (SCI) and are believed to entail distinct pathways; nevertheless, they may share shared defective glial systems. Neuropathic pain after SCI is caused by glial cells, ectopic firing of neurons endings and their intra- and extracellular signaling mechanisms. One such mechanism occurs when stimuli that were previously non-noxious become so after the injury. This will exhibit a symptom of allodynia. Another mechanism is the release of substances by glia, which keeps the sensitivity of dorsal horn neurons even in regions distant from the site of injury. Here, we review, the models and identifications of SCI-induced neuropathic pain (SCI-NP), the mechanisms of SCI-NP related to glia, and the treatments of SCI-NP.
Collapse
Affiliation(s)
- Ying Ye
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinjin Su
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Tang
- Department of Anesthesiology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, China
| | - Chao Zhu
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
45
|
Castro E Silva JH, Pieropan F, Rivera AD, Butt AM, Costa SL. Agathisflavone Modulates Reactive Gliosis After Trauma and Increases the Neuroblast Population at the Subventricular Zone. Nutrients 2024; 16:4053. [PMID: 39683446 DOI: 10.3390/nu16234053] [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: 11/09/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Reactive astrogliosis and microgliosis are coordinated responses to CNS insults and are pathological hallmarks of traumatic brain injury (TBI). In these conditions, persistent reactive gliosis can impede tissue repopulation and limit neurogenesis. Thus, modulating this phenomenon has been increasingly recognized as potential therapeutic approach. METHODS In this study, we investigated the potential of the flavonoid agathisflavone to modulate astroglial and microglial injury responses and promote neurogenesis in the subventricular zone (SVZ) neurogenic niche. Agathisflavone, or the vehicle in controls, was administered directly into the lateral ventricles in postnatal day (P)8-10 mice by twice daily intracerebroventricular (ICV) injections for 3 days, and brains were examined at P11. RESULTS In the controls, ICV injection caused glial reactivity along the needle track, characterised immunohistochemically by increased astrocyte expression of glial fibrillary protein (GFAP) and the number of Iba-1+ microglia at the lesion site. Treatment with agathisflavone decreased GFAP expression, reduced both astrocyte reactivity and the number of Iba-1+ microglia at the core of the lesion site and the penumbra, and induced a 2-fold increase on the ratio of anti-inflammatory CD206+ to pro-inflammatory CD16/32+ microglia. Notably, agathisflavone increased the population of neuroblasts (GFAP+ type B cells) in all SVZ microdomains by up to double, without significantly increasing the number of neuronal progenitors (DCX+). CONCLUSIONS Although future studies should investigate the underlying molecular mechanisms driving agathisflavone effects on microglial polarization and neurogenesis at different timepoints, these data indicate that agathisflavone could be a potential adjuvant treatment for TBI or central nervous system disorders that have reactive gliosis as a common feature.
Collapse
Affiliation(s)
- Juliana Helena Castro E Silva
- Laboratory of Neurochemistry and Cellular Biology, Department of Biofunction, Health Sciences Institute, Federal University of Bahia, Salvador 40231-300, Brazil
| | - Francesca Pieropan
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK
- Southampton Solent University, E Park Terrace, Southampton SO14 0YN, UK
| | - Andrea Domenico Rivera
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK
| | - Arthur Morgan Butt
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Department of Biofunction, Health Sciences Institute, Federal University of Bahia, Salvador 40231-300, Brazil
| |
Collapse
|
46
|
Bhatt M, Sharma M, Das B. The Role of Inflammatory Cascade and Reactive Astrogliosis in Glial Scar Formation Post-spinal Cord Injury. Cell Mol Neurobiol 2024; 44:78. [PMID: 39579235 PMCID: PMC11585509 DOI: 10.1007/s10571-024-01519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024]
Abstract
Reactive astrogliosis and inflammation are pathologic hallmarks of spinal cord injury. After injury, dysfunction of glial cells (astrocytes) results in glial scar formation, which limits neuronal regeneration. The blood-spinal cord barrier maintains the structural and functional integrity of the spinal cord and does not allow blood vessel components to leak into the spinal cord microenvironment. After the injury, disruption in the spinal cord barrier causes an imbalance of the immunological microenvironment. This triggers the process of neuroinflammation, facilitated by the actions of microglia, neutrophils, glial cells, and cytokines production. Recent work has revealed two phenotypes of astrocytes, A1 and A2, where A2 has a protective type, and A1 releases neurotoxins, further promoting glial scar formation. Here, we first describe the current understanding of the spinal cord microenvironment, both pre-, and post-injury, and the role of different glial cells in the context of spinal cord injury, which forms the essential update on the cellular and molecular events following injury. We aim to explore in-depth signaling pathways and molecular mediators that trigger astrocyte activation and glial scar formation. This review will discuss the activated signaling pathways in astrocytes and other glial cells and their collaborative role in the development of gliosis through inflammatory responses.
Collapse
Affiliation(s)
- Manini Bhatt
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Muskan Sharma
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, India.
| |
Collapse
|
47
|
Jian H, Wu K, Lv Y, Du J, Hou M, Zhang C, Gao J, Zhou H, Feng S. A critical role for microglia in regulating metabolic homeostasis and neural repair after spinal cord injury. Free Radic Biol Med 2024; 225:469-481. [PMID: 39413980 DOI: 10.1016/j.freeradbiomed.2024.10.288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Traumatic spinal cord injury (SCI) often results in severe immune and metabolic disorders, aggravating neurological damage and inhibiting locomotor functional recovery. Microglia, as resident immune cells of the spinal cord, play crucial roles in maintaining neural homeostasis under physiological conditions. However, the precise role of microglia in regulating immune and metabolic functions in SCI is still unclear and is easily confused with that of macrophages. In this study, we pharmacologically depleted microglia to explore the role of microglia after SCI. We found that microglia are beneficial for the recovery of locomotor function. Depleting microglia disrupted glial scar formation, reducing neurogenesis and angiogenesis. Using liquid chromatography tandem mass spectrometry (LC‒MS/MS), we discovered that depleting microglia significantly inhibits lipid metabolism processes such as fatty acid degradation, unsaturated fatty acid biosynthesis, glycophospholipid metabolism, and sphingolipid metabolism, accompanied by the accumulation of multiple organic acids. Subsequent studies demonstrated that microglial depletion increased the inhibition of FASN after SCI. FASN inhibition exacerbated malonyl-CoA accumulation and significantly impeded the activity of mTORC1. Moreover, microglial depletion exacerbated the oxidative stress of neurons. In summary, our results indicate that microglia alleviate neural damage and metabolic disorders after SCI, which is beneficial for achieving optimal neuroprotection and neural repair.
Collapse
Affiliation(s)
- Huan Jian
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Kailin Wu
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Yigang Lv
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Jiawei Du
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Mengfan Hou
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China
| | - Chi Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Hengxing Zhou
- Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Department of Orthopaedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin, China; Department of Orthopaedics, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Shandong University Center for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Orthopedics, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
48
|
Zucha D, Abaffy P, Kirdajova D, Jirak D, Kubista M, Anderova M, Valihrach L. Spatiotemporal transcriptomic map of glial cell response in a mouse model of acute brain ischemia. Proc Natl Acad Sci U S A 2024; 121:e2404203121. [PMID: 39499634 PMCID: PMC11573666 DOI: 10.1073/pnas.2404203121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024] Open
Abstract
The role of nonneuronal cells in the resolution of cerebral ischemia remains to be fully understood. To decode key molecular and cellular processes that occur after ischemia, we performed spatial and single-cell transcriptomic profiling of the male mouse brain during the first week of injury. Cortical gene expression was severely disrupted, defined by inflammation and cell death in the lesion core, and glial scar formation orchestrated by multiple cell types on the periphery. The glial scar was identified as a zone with intense cell-cell communication, with prominent ApoE-Trem2 signaling pathway modulating microglial activation. For each of the three major glial populations, an inflammatory-responsive state, resembling the reactive states observed in neurodegenerative contexts, was observed. The recovered spectrum of ischemia-induced oligodendrocyte states supports the emerging hypothesis that oligodendrocytes actively respond to and modulate the neuroinflammatory stimulus. The findings are further supported by analysis of other spatial transcriptomic datasets from different mouse models of ischemic brain injury. Collectively, we present a landmark transcriptomic dataset accompanied by interactive visualization that provides a comprehensive view of spatiotemporal organization of processes in the postischemic mouse brain.
Collapse
Affiliation(s)
- Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
- Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology, Prague 16000, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| | - Denisa Kirdajova
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Daniel Jirak
- Department of Radiodiagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague 14021, Czech Republic
- Faculty of Health Studies, Technical University of Liberec, Liberec 46001, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec 25250, Czech Republic
| |
Collapse
|
49
|
Andrew PM, MacMahon JA, Bernardino PN, Tsai YH, Hobson BA, Porter VA, Huddleston SL, Luo AS, Bruun DA, Saito NH, Harvey DJ, Brooks-Kayal A, Chaudhari AJ, Lein PJ. Shifts in the spatiotemporal profile of inflammatory phenotypes of innate immune cells in the rat brain following acute intoxication with the organophosphate diisopropylfluorophosphate. J Neuroinflammation 2024; 21:285. [PMID: 39497181 PMCID: PMC11533402 DOI: 10.1186/s12974-024-03272-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Acute intoxication with cholinesterase inhibiting organophosphates (OP) can produce life-threatening cholinergic crisis and status epilepticus (SE). Survivors often develop long-term neurological consequences, including spontaneous recurrent seizures (SRS) and impaired cognition. Numerous studies implicate OP-induced neuroinflammation as a pathogenic mechanism contributing to these chronic sequelae; however, little is known about the inflammatory phenotype of innate immune cells in the brain following acute OP intoxication. Thus, the aim of this study was to characterize the natural history of microglial and astrocytic inflammatory phenotypes following acute intoxication with the OP, diisopropylfluorophosphate (DFP). Adult male and female Sprague-Dawley rats were administered a single dose of DFP (4 mg/kg, sc) followed by standard medical countermeasures. Within minutes, animals developed benzodiazepine-resistant SE as determined by monitoring seizures using a modified Racine scale. At 1, 3, 7, 14, and 28 d post-exposure (DPE), neuroinflammation was assessed using translocator protein (TSPO) positron emission tomography (PET) and magnetic resonance imaging (MRI). In both sexes, we observed consistently elevated radiotracer uptake across all examined brain regions and time points. A separate group of animals was euthanized at these same time points to collect tissues for immunohistochemical analyses. Colocalization of IBA-1, a marker for microglia, with iNOS or Arg1 was used to identify pro- and anti-inflammatory microglia, respectively; colocalization of GFAP, a marker for astrocytes, with C3 or S100A10, pro- and anti-inflammatory astrocytes, respectively. We observed shifts in the inflammatory profiles of microglia and astrocyte populations during the first month post-intoxication, largely in hyperintense inflammatory lesions in the piriform cortex and amygdala regions. In these areas, iNOS+ proinflammatory microglial cell density peaked at 3 and 7 DPE, while anti-inflammatory Arg1+ microglia cell density peaked at 14 DPE. Pro- and anti-inflammatory astrocytes emerged within 7 DPE, and roughly equal ratios of C3+ pro-inflammatory and S100A10+ anti-inflammatory astrocytes persisted at 28 DPE. In summary, microglia and astrocytes adopted mixed inflammatory phenotypes post-OP intoxication, which evolved over one month post exposure. These activated cell populations were most prominent in the piriform and amygdala areas and were more abundant in males compared to females. The temporal relationship between microglial and astrocytic responses suggests that initial microglial activity may influence delayed, persistent astrocytic responses. Further, our findings identify putative windows for inhibition of OP-induced neuroinflammatory responses in both sexes to evaluate the therapeutic benefit of anti-inflammation in this context.
Collapse
Affiliation(s)
- Peter M Andrew
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Jeremy A MacMahon
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Pedro N Bernardino
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Yi-Hua Tsai
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Brad A Hobson
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Valerie A Porter
- Department of Biomedical Engineering, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Sydney L Huddleston
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Audrey S Luo
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Donald A Bruun
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Naomi H Saito
- Department of Public Health Sciences, Davis, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Danielle J Harvey
- Department of Public Health Sciences, Davis, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Amy Brooks-Kayal
- Department of Neurology, Davis, School of Medicine, University of California, Sacramento, CA, 95817, USA
| | - Abhijit J Chaudhari
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
- Department of Radiology, Davis, School of Medicine, University of California, Sacramento, CA, 95817, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
50
|
Huang Y, Bai J. Ferroptosis in the neurovascular unit after spinal cord injury. Exp Neurol 2024; 381:114943. [PMID: 39242069 DOI: 10.1016/j.expneurol.2024.114943] [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: 06/10/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The mechanisms of secondary injury following spinal cord injury are complicated. The role of ferroptosis, which is a newly discovered form of regulated cell death in the neurovascular unit(NVU), is increasingly important. Ferroptosis inhibitors have been shown to improve neurovascular homeostasis and attenuate secondary spinal cord injury(SCI). This review focuses on the mechanisms of ferroptosis in NVU cells and NVU-targeted therapeutic strategies according to the stages of SCI, and analyzes possible future research directions.
Collapse
Affiliation(s)
- Yushan Huang
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Jinzhu Bai
- School of Rehabilitation, Capital Medical University, Beijing, China; Department of Spine and Spinal Cord Surgery, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing, China; Department of Orthopedics, Capital Medical University, Beijing, China.
| |
Collapse
|