1
|
Avonts BL, Shen Q, Wrobel NJ, Fessler RG, David BT. The relationship between changes in inflammation and locomotor function in sensory phenotypes of central neuropathic pain after spinal cord injury. Pain Rep 2024; 9:e1184. [PMID: 39399305 PMCID: PMC11469887 DOI: 10.1097/pr9.0000000000001184] [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: 12/08/2023] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Central neuropathic pain (CNP) commonly develops in patients after spinal cord injury (SCI), causing debilitating symptoms and sensory abnormalities to mechanical and thermal stimuli. The biological variability of pain phenotypes in individuals has limited the number of positive outcomes. Thus, it is necessary to investigate the physiological processes contributing to sensory changes that develop over time. Objective To investigate the physiological processes contributing to neuropathic pain sensory changes and locomotor impairments with sensory phenotypes that develop over time. Methods Using the tail flick and von Frey tests, we performed hierarchical clustering to determine the subpopulation of rats that developed thermal and mechanical sensory abnormalities. To measure inflammation as a potential mediator of CNP phenotypes, we used flow cytometry and immunohistochemistry. Finally, to assess the secondary effects on locomotor recovery, up to 8 weeks after injury, we used the CatWalk test to assess multiple parameters of gait. Results The von Frey test showed a subpopulation of SCI rats that were hyposensitive to mechanical stimuli from 6 to 8 weeks after injury. The tail flick test showed a subpopulation of SCI rats that were hypersensitive to thermal stimuli at 1 week and 3 to 8 weeks after injury. Although there were no differences in inflammatory cells between subpopulations, we did see significant changes in locomotor recovery between rats with and without sensory abnormalities. Conclusion The myeloid cell population at large is not affected by mechanical or thermal phenotypes of pain in this model; however, locomotor recovery is impaired depending on the pain phenotype present. Further investigation into acute inflammatory cells may be insightful for predicting the development of pain phenotypes.
Collapse
Affiliation(s)
- Brittany L. Avonts
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Quan Shen
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Neal J. Wrobel
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Richard G. Fessler
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| | - Brian T. David
- Rush University Medical Center, Department of Neurosurgery, Chicago, IL, USA
| |
Collapse
|
2
|
Cao J, Yu X, Liu J, Fu J, Wang B, Wu C, Zhang S, Chen H, Wang Z, Xu Y, Sui T, Chang J, Cao X. Ruxolitinib improves the inflammatory microenvironment, restores glutamate homeostasis, and promotes functional recovery after spinal cord injury. Neural Regen Res 2024; 19:2499-2512. [PMID: 38526286 PMCID: PMC11090442 DOI: 10.4103/nrr.nrr-d-23-01863] [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: 11/12/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00030/figure1/v/2024-03-08T184507Z/r/image-tiff The inflammatory microenvironment and neurotoxicity can hinder neuronal regeneration and functional recovery after spinal cord injury. Ruxolitinib, a JAK-STAT inhibitor, exhibits effectiveness in autoimmune diseases, arthritis, and managing inflammatory cytokine storms. Although studies have shown the neuroprotective potential of ruxolitinib in neurological trauma, the exact mechanism by which it enhances functional recovery after spinal cord injury, particularly its effect on astrocytes, remains unclear. To address this gap, we established a mouse model of T10 spinal cord contusion and found that ruxolitinib effectively improved hindlimb motor function and reduced the area of spinal cord injury. Transcriptome sequencing analysis showed that ruxolitinib alleviated inflammation and immune response after spinal cord injury, restored EAAT2 expression, reduced glutamate levels, and alleviated excitatory toxicity. Furthermore, ruxolitinib inhibited the phosphorylation of JAK2 and STAT3 in the injured spinal cord and decreased the phosphorylation level of nuclear factor kappa-B and the expression of inflammatory factors interleukin-1β, interleukin-6, and tumor necrosis factor-α. Additionally, in glutamate-induced excitotoxicity astrocytes, ruxolitinib restored EAAT2 expression and increased glutamate uptake by inhibiting the activation of STAT3, thereby reducing glutamate-induced neurotoxicity, calcium influx, oxidative stress, and cell apoptosis, and increasing the complexity of dendritic branching. Collectively, these results indicate that ruxolitinib restores glutamate homeostasis by rescuing the expression of EAAT2 in astrocytes, reduces neurotoxicity, and effectively alleviates inflammatory and immune responses after spinal cord injury, thereby promoting functional recovery after spinal cord injury.
Collapse
Affiliation(s)
- Jiang Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiao Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jingcheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiaju Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Binyu Wang
- Department of Trauma Surgery, Subei People’s Hospital of Jiangsu, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Chaoqin Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sheng Zhang
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Hongtao Chen
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Zi Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yinyang Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Sui
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Chang
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
3
|
Zhang L, Wei J, Huang Y, Wang L, Gao H, Yang Y. Clickable immune-microenvironment modulated hydrogels for spinal cord injury repair. J Colloid Interface Sci 2024; 679:1079-1092. [PMID: 39504844 DOI: 10.1016/j.jcis.2024.10.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/07/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024]
Abstract
Spinal cord injury (SCI) is a devastating condition without effective therapy currently available. The inflammatory cascade following SCI leads to neuronal apoptosis and glial cell activation. The utilization of local injectable hydrogels with immunotherapy drugs directly into injured nerve tissues represents a promising therapeutic strategy. Herein, injectable hydrogels grafted with clickable methylprednisolone (MP) and cellular adhesion peptide were developed using free radical polymerization for promoting nerve regeneration following SCI. MP conjugated hydrogels could modulate the immunoinflammatory microenvironment of SCI and sustain neuron survival. The multi-stiffness hydrogels were fabricated by adjusting concentration ratios to evaluate appropriate mechanical stimuli. In a model of dorsal root ganglion, MP grafted hydrogels with mechanical signals similar to those of adult rat spinal cords demonstrated superior efficacy in promoting dorsal root ganglion growth. MP grafted hydrogels could regulate the immune-inflammatory microenvironment, promote recovery of both motor function and sensory functions. The positive findings suggested that the interplay between immunomodulation and mechanical signals plays a crucial role in promoting nerve regeneration, indicating significant potential for hydrogels as a therapeutic approach for repairing SCI.
Collapse
Affiliation(s)
- Luzhong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, PR China
| | - Jingjing Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, PR China
| | - Yuan Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, PR China
| | - Luqi Wang
- Machinery and Electronics Engineering College, Shandong Agriculture and Engineering University, PR China
| | - Huasong Gao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong 226001, PR China.
| |
Collapse
|
4
|
Shan F, Ye J, Xu X, Liang C, Zhao Y, Wang J, Ouyang F, Li J, Lv J, Wu Z, Yao F, Jing J, Zheng M. Galectin-3 inhibition reduces fibrotic scarring and promotes functional recovery after spinal cord injury in mice. Cell Biosci 2024; 14:128. [PMID: 39407295 PMCID: PMC11481377 DOI: 10.1186/s13578-024-01310-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND In the context of spinal cord injury (SCI), infiltrating macrophages assume prominence as the primary inflammatory cells within the lesion core, where the fibrotic scar is predominantly orchestrated by platelet-derived growth factor receptor beta (PDGFRβ+) fibroblasts. Galectin-3, a carbohydrate-binding protein of the lectin family, is notably expressed by infiltrating hematogenous macrophages and mediates cell-cell interactions. Although Galectin-3 has been shown to contribute to the endocytic internalization of PDGFRβ in vitro, its specific role in driving fibrotic scar formation after SCI has not been determined. METHODS We employed a crush mid-thoracic (T10) SCI mouse model. Galectin-3 inhibition after SCI was achieved through intrathecal injection of the Galectin-3 inhibitor TD139 or in situ injection of lentivirus carrying Galectin-3-shRNA (Lv-shLgals3). A fibrosis-induced mice model was established by in situ injection of platelet-derived growth factor D (PDGFD) or recombinant Galectin-3 (rGalectin-3) into the uninjured spinal cord. Galectin-3 internalization experiments were conducted in PDGFRβ+ fibroblasts cocultured in conditioned medium in vitro. RESULTS We identified the spatial and temporal correlation between macrophage-derived Galectin-3 and PDGFRβ in fibroblasts from 3 to 56 days post-injury (dpi). Administration of TD139 via intrathecal injection or in situ injection of Lv-shLgals3 effectively mitigated fibrotic scar formation and extracellular matrix deposition within the injured spinal cord, leading to better neurological outcomes and function recovery after SCI. Furthermore, the fibrosis-inducing effects of exogenous PDGFD in the uninjured spinal cord could be blocked by TD139. In vitro experiments further demonstrated the ability of PDGFRβ+ fibroblasts to internalize Galectin-3, with Galectin-3 inhibition resulting in reduced PDGFRβ expression. CONCLUSIONS Our finding underscores the pivotal role of macrophage-derived Galectin-3 in modulating the sustained internalized activation of PDGFRβ within fibroblasts, providing a novel mechanistic insight into fibrotic scarring post-SCI.
Collapse
Affiliation(s)
- Fangli Shan
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jianan Ye
- 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
- Department of Orthopaedics, Suzhou 100 Hospital, Suzhou, 215000, China
| | - Xinzhong Xu
- Department of Orthopaedics, 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
| | - Yuanzhe Zhao
- 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
| | - Fangru Ouyang
- 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
| | - Jianwei Lv
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhonghan Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Fei Yao
- 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
|
5
|
Shi Y, Zheng M, Luo Y, Li J, Ouyang F, Zhao Y, Wang J, Ma Z, Meng C, Bi Y, Cheng L, Jing J. Targeting transcription factor pu.1 for improving neurologic outcomes after spinal cord injury. Front Neurosci 2024; 18:1418615. [PMID: 39211434 PMCID: PMC11358095 DOI: 10.3389/fnins.2024.1418615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/10/2024] [Indexed: 09/04/2024] Open
Abstract
Background After spinal cord injury (SCI), lipid metabolism dysregulation at the lesion site exacerbates secondary damage. The transcription factor pu.1 has been implicated as a negative regulator of multiple lipid metabolism-related genes and pathways. However, its role in post-SCI lipid metabolism remains unclear. Methods We employed a mouse model of complete T10 crush SCI. Non-targeted metabolomics and bioinformatics analysis were utilized to investigate lipid metabolism at the lesion site after SCI. Polarized light imaging was used to evaluate the presence of cholesterol crystals. DB1976, a specific inhibitor of pu.1, was administered to examine its impact on local lipid metabolism after SCI. Immunofluorescence staining was performed to assess pu.1 expression and distribution, and to evaluate lipid droplet formation, astrocytic/fibrotic scar development, inflammatory cell infiltration, and tight junctions within the vasculature. Results Non-targeted metabolomics and bioinformatics analyses revealed significant alterations in lipid metabolism components after SCI. Moreover, immunofluorescence staining and polarized light imaging demonstrated substantial BODIPY+ lipid droplet accumulation and persistent cholesterol crystal formation at the lesion site after SCI. Increased pu.1 expression was predominantly observed within macrophages/microglia at the lesion site after SCI. DB1976 treatment significantly mitigated lipid droplet accumulation and cholesterol crystal formation, reduced CD68+ macrophage/microglial infiltration, and attenuated fibrotic scar formation. Moreover, DB1976 treatment promoted the expression of claudin-5 and zonula occludens-1 between vascular endothelial cells and enhanced GFAP+ glial connectivity after SCI. Conclusion Our study reveals a significant correlation between lipid metabolism disturbance post-SCI and transcription factor pu.1 upregulation, specifically in macrophages/microglia at the lesion site. Thus, targeted pu.1 modulation has the potential to yield promising results by substantially diminishing the deposition of lipid metabolism byproducts at the lesion site and fostering a milieu conducive to SCI repair.
Collapse
Affiliation(s)
- Yi Shi
- 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
| | - Yang Luo
- 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
| | - 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
| | - 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
| | - Yuanzhe Zhao
- 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
| | - Jingwen Wang
- 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
| | - Zhida Ma
- 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
| | - Congpeng Meng
- 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
| | - Yihui Bi
- 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
|
Li Z, Zhou T, Bao Z, Wu M, Mao Y. The Porous SilMA Hydrogel Scaffolds Carrying Dual-Sensitive Paclitaxel Nanoparticles Promote Neuronal Differentiation for Spinal Cord Injury Repair. Tissue Eng Regen Med 2024; 21:809-827. [PMID: 39004636 PMCID: PMC11286913 DOI: 10.1007/s13770-024-00659-9] [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: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND In the intricate pathological milieu post-spinal cord injury (SCI), neural stem cells (NSCs) frequently differentiate into astrocytes rather than neurons, significantly limiting nerve repair. Hence, the utilization of biocompatible hydrogel scaffolds in conjunction with exogenous factors to foster the differentiation of NSCs into neurons has the potential for SCI repair. METHODS In this study, we engineered a 3D-printed porous SilMA hydrogel scaffold (SM) supplemented with pH-/temperature-responsive paclitaxel nanoparticles (PTX-NPs). We analyzed the biocompatibility of a specific concentration of PTX-NPs and its effect on NSC differentiation. We also established an SCI model to explore the ability of composite scaffolds for in vivo nerve repair. RESULTS The physical adsorption of an optimal PTX-NPs dosage can simultaneously achieve pH/temperature-responsive release and commendable biocompatibility, primarily reflected in cell viability, morphology, and proliferation. An appropriate PTX-NPs concentration can steer NSC differentiation towards neurons over astrocytes, a phenomenon that is also efficacious in simulated injury settings. Immunoblotting analysis confirmed that PTX-NPs-induced NSC differentiation occurred via the MAPK/ERK signaling cascade. The repair of hemisected SCI in rats demonstrated that the composite scaffold augmented neuronal regeneration at the injury site, curtailed astrocyte and fibrotic scar production, and enhanced motor function recovery in rat hind limbs. CONCLUSION The scaffold's porous architecture serves as a cellular and drug carrier, providing a favorable microenvironment for nerve regeneration. These findings corroborate that this strategy amplifies neuronal expression within the injury milieu, significantly aiding in SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Tao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Zhengqi Bao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China.
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
- Anhui Engineering Research Center of Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
7
|
Wang L, Zhao H, Han M, Yang H, Lei M, Wang W, Li K, Li Y, Sang Y, Xin T, Liu H, Qiu J. Electromagnetic Cellularized Patch with Wirelessly Electrical Stimulation for Promoting Neuronal Differentiation and Spinal Cord Injury Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307527. [PMID: 38868910 PMCID: PMC11321663 DOI: 10.1002/advs.202307527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/02/2024] [Indexed: 06/14/2024]
Abstract
Although stem cell therapy holds promise for the treatment of spinal cord injury (SCI), its practical applications are limited by the low degree of neural differentiation. Electrical stimulation is one of the most effective ways to promote the differentiation of stem cells into neurons, but conventional wired electrical stimulation may cause secondary injuries, inflammation, pain, and infection. Here, based on the high conductivity of graphite and the electromagnetic induction effect, graphite nanosheets with neural stem cells (NSCs) are proposed as an electromagnetic cellularized patch to generate in situ wirelessly pulsed electric signals under a rotating magnetic field for regulating neuronal differentiation of NSCs to treat SCI. The strength and frequency of the induced voltage can be controlled by adjusting the rotation speed of the magnetic field. The generated pulsed electrical signals promote the differentiation of NSCs into functional mature neurons and increase the proportion of neurons from 12.5% to 33.7%. When implanted in the subarachnoid region of the injured spinal cord, the electromagnetic cellularized patch improves the behavioral performance of the hind limbs and the repair of spinal cord tissue in SCI mice. This work opens a new avenue for remote treatment of SCI and other nervous system diseases.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Hongbo Zhao
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
| | - Min Han
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
| | - Hongru Yang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Ming Lei
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Wenhan Wang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Keyi Li
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Yiwei Li
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Yuanhua Sang
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| | - Tao Xin
- Department of NeurosurgeryThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinan250014P. R. China
- Department of Neurosurgery, Shandong Provincial Qianfoshan HospitalShandong UniversityJinan250014P. R. China
- Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinan250117P. R. China
- Department of NeurosurgeryJiangxi Provincial People's HospitalNanchangJiangxi330006P. R. China
| | - Hong Liu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
- Institute for Advanced Interdisciplinary ResearchUniversity of JinanJinanShandong250022P. R. China
| | - Jichuan Qiu
- State Key Laboratory of Crystal MaterialsShandong UniversityJinanShandong250100P. R. China
| |
Collapse
|
8
|
Gaire S, An J, Yang H, Lee KA, Dumre M, Lee EJ, Park SM, Joe EH. Systemic inflammation attenuates the repair of damaged brains through reduced phagocytic activity of monocytes infiltrating the brain. Mol Brain 2024; 17:47. [PMID: 39075534 PMCID: PMC11288066 DOI: 10.1186/s13041-024-01116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/19/2024] [Indexed: 07/31/2024] Open
Abstract
In this study, we examined how systemic inflammation affects repair of brain injury. To this end, we created a brain-injury model by stereotaxic injection of ATP, a damage-associated molecular pattern component, into the striatum of mice. Systemic inflammation was induced by intraperitoneal injection of lipopolysaccharide (LPS-ip). An analysis of magnetic resonance images showed that LPS-ip reduced the initial brain injury but slowed injury repair. An immunostaining analysis using the neuronal marker, NeuN, showed that LPS-ip delayed removal of dead/dying neurons, despite the fact that LPS-ip enhanced infiltration of monocytes, which serve to phagocytize dead cells/debris. Notably, infiltrating monocytes showed a widely scattered distribution. Bulk RNAseq analyses showed that LPS-ip decreased expression of genes associated with phagocytosis, with PCR and immunostaining of injured brains confirming reduced levels of Cd68 and Clec7a, markers of phagocytic activity, in monocytes. Collectively, these results suggest that systemic inflammation affects properties of blood monocytes as well as brain cells, resulting in delay in clearing damaged cells and activating repair processes.
Collapse
Affiliation(s)
- Sushil Gaire
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Jiawei An
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Haijie Yang
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Keon Ah Lee
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Manisha Dumre
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Eun Jeong Lee
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Sang-Myun Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.
- Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, South Korea.
| |
Collapse
|
9
|
Lin K, Zhang Y, Shen Y, Xu Y, Huang M, Liu X. Hydrogen Sulfide can Scavenge Free Radicals to Improve Spinal Cord Injury by Inhibiting the p38MAPK/mTOR/NF-κB Signaling Pathway. Neuromolecular Med 2024; 26:26. [PMID: 38907170 DOI: 10.1007/s12017-024-08794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.
Collapse
Affiliation(s)
- Kexin Lin
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yanyang Shen
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yiqin Xu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
10
|
Huang J, Hu X, Chen Z, Ouyang F, Li J, Hu Y, Zhao Y, Wang J, Yao F, Jing J, Cheng L. Fascin-1 limits myosin activity in microglia to control mechanical characterization of the injured spinal cord. J Neuroinflammation 2024; 21:88. [PMID: 38600569 PMCID: PMC11005239 DOI: 10.1186/s12974-024-03089-5] [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: 01/26/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Mechanical softening of the glial scar region regulates axonal regeneration to impede neurological recovery in central nervous system (CNS) injury. Microglia, a crucial cellular component of the glial scar, facilitate neuronal survival and neurological recovery after spinal cord injury (SCI). However, the critical mechanical characterization of injured spinal cord that harmonizes neuroprotective function of microglia remains poorly understood. METHODS Spinal cord tissue stiffness was assessed using atomic force microscopy (AFM) in a mouse model of crush injury. Pharmacological depletion of microglia using PLX5622 was used to explore the effect of microglia on mechanical characterization. Conditional knockout of Fascin-1 in microglia (Fascin-1 CKO) alone or in combination with inhibition of myosin activity was performed to delve into relevant mechanisms of microglia regulating mechanical signal. Immunofluorescence staining was performed to evaluate the related protein levels, inflammatory cells, and neuron survival after SCI. The Basso mouse scale score was calculated to assess functional recovery. RESULTS Spinal cord tissue significantly softens after SCI. Microglia depletion or Fascin-1 knockout in microglia limits tissue softening and alters mechanical characterization, which leads to increased tissue pathology and impaired functional recovery. Mechanistically, Fascin-1 inhibits myosin activation to promote microglial migration and control mechanical characterization after SCI. CONCLUSIONS We reveal that Fascin-1 limits myosin activity to regulate mechanical characterization after SCI, and this mechanical signal should be considered in future approaches for the treatment of CNS diseases.
Collapse
Affiliation(s)
- Jinxin Huang
- 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
| | - Xuyang Hu
- 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
| | - Zeqiang Chen
- 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
| | - Fangru Ouyang
- 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
| | - Jianjian 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
| | - Yixue Hu
- 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
| | - Yuanzhe Zhao
- 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
| | - Jingwen Wang
- 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
| | - Fei Yao
- 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.
| | - Li Cheng
- 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
|
11
|
Romenskaja D, Jonavičė U, Tunaitis V, Pivoriūnas A. Extracellular vesicles from oral mucosa stem cells promote lipid raft formation in human microglia through TLR4, P2X4R, and αVβ3/αVβ5 signaling pathways. Cell Biol Int 2024; 48:358-368. [PMID: 38100213 DOI: 10.1002/cbin.12111] [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: 06/14/2023] [Revised: 09/24/2023] [Accepted: 12/01/2023] [Indexed: 02/15/2024]
Abstract
Targeting of disease-associated microglia represents a promising therapeutic approach that can be used for the prevention or slowing down neurodegeneration. In this regard, the use of extracellular vesicles (EVs) represents a promising therapeutic approach. However, the molecular mechanisms by which EVs regulate microglial responses remain poorly understood. In the present study, we used EVs derived from human oral mucosa stem cells (OMSCs) to investigate the effects on the lipid raft formation and the phagocytic response of human microglial cells. Lipid raft labeling with fluorescent cholera toxin subunit B conjugates revealed that both EVs and lipopolysaccharide (LPS) by more than two times increased lipid raft formation in human microglia. By contrast, combined treatment with LPS and EVs significantly decreased lipid raft formation indicating possible interference of EVs with the process of LPS-induced lipid raft formation. Specific inhibition of Toll-like receptor 4 (TLR4) with anti-TLR4 antibody as well as inhibition of purinergic P2X4 receptor (P2X4R) with selective antagonist 5-BDBD inhibited EVs- and LPS-induced lipid raft formation. Selective blockage of αvβ3/αvβ5 integrins with cilengitide suppressed EV- and LPS-induced lipid raft formation in microglia. Furthermore, inhibition of TLR4 and P2X4R prevented EV-induced phagocytic activity of human microglial cells. We demonstrate that EVs induce lipid raft formation in human microglia through interaction with TLR4, P2X4R, and αVβ3/αVβ5 signaling pathways. Our results provide new insights about the molecular mechanisms regulating EV/microglia interactions and could be used for the development of new therapeutic strategies against neurological disorders.
Collapse
Affiliation(s)
- Diana Romenskaja
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ugnė Jonavičė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Virginijus Tunaitis
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
12
|
Shen K, Li X, Huang G, Yuan Z, Xie B, Chen T, He L. High rapamycin-loaded hollow mesoporous Prussian blue nanozyme targets lesion area of spinal cord injury to recover locomotor function. Biomaterials 2023; 303:122358. [PMID: 37951099 DOI: 10.1016/j.biomaterials.2023.122358] [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: 04/11/2023] [Revised: 09/09/2023] [Accepted: 10/17/2023] [Indexed: 11/13/2023]
Abstract
Scavenging free radicals and reducing inflammatory reaction to relieve the secondary damage are important issues in the spinal cord injury (SCI) therapeutic strategy. Nanozymes attract more attention in the drug development of SCI due to the high stability, long-lasting catalytic capacity, and multienzyme-like properties. Herein, we constructed a Rapamycin (Rapa)-loaded and hollow mesoporous Prussian blue (HMPB)-based nanozyme (RHPAzyme) to realize the combined antioxidation and anti-inflammation combination therapy of SCI. Furthermore, activated cell penetrating peptide (ACPP) is modified onto nanozyme to endow the effectively ability of lesion area-targeting. This RHPAzyme exhibits ROS scavenging capacity with the transformation of Fe2+/Fe3+ valance and cyanide group of HMPB to achieve multienzyme-like activity. As expected, RHPAzyme scavenges the ROS overproduction and reduces inflammation in oxygen-glucose deprivation (OGD)-induced damage via inhibiting MAPK/AKT signaling pathway. Furtherly, RHPAzyme exhibits the combined antioxidant and anti-inflammatory activity in vivo, which can effectively alleviate neuronal damage and promote motor function recovery in SCI mice. Overall, this study demonstrates the RHPAzyme induces an effective treatment of SCI by inhibiting oxygen-mediated cell apoptosis and suppressing inflammation-induced injury, thus reduces the nervous impairment and promotes motor function recovery.
Collapse
Affiliation(s)
- Kui Shen
- Department of Orthopedics, Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaowei Li
- Department of Orthopedics, Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Guanning Huang
- Department of Orthopedics, Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China; Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Zhongwen Yuan
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Bin Xie
- Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- Department of Orthopedics, Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Lizhen He
- Department of Orthopedics, Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510632, China.
| |
Collapse
|
13
|
Kitade K, Kobayakawa K, Saiwai H, Matsumoto Y, Kawaguchi K, Iida K, Kijima K, Iura H, Tamaru T, Haruta Y, Ono G, Konno D, Maeda T, Okada S, Nakashima K, Nakashima Y. Reduced Neuroinflammation Via Astrocytes and Neutrophils Promotes Regeneration After Spinal Cord Injury in Neonatal Mice. J Neurotrauma 2023; 40:2566-2579. [PMID: 37503626 DOI: 10.1089/neu.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Neonatal spinal cord injury (SCI) shows better functional outcomes than adult SCI. Although the regenerative capability in the neonatal spinal cord may have cues in the treatment of adult SCI, the mechanism underlying neonatal spinal cord regeneration after SCI is unclear. We previously reported age-dependent variation in the pathogenesis of inflammation after SCI. Therefore, we explored differences in the pathogenesis of inflammation after SCI between neonatal and adult mice and their effect on axon regeneration and functional outcome. We established two-day-old spinal cord crush mice as a model of neonatal SCI. Immunohistochemistry of the spinal cord revealed that the nuclear translocation of NF-κB, which promotes the expression of chemokines, was significantly lower in the astrocytes of neonates than in those of adults. Flow cytometry revealed that neonatal astrocytes secrete low levels of chemokines to recruit circulating neutrophils (e.g., Cxcl1 and Cxcl2) after SCI in comparison with adults. We also found that the expression of a chemokine receptor (CXCR2) and an adhesion molecule (β2 integrin) quantified by flow cytometry was lower in neonatal circulating neutrophils than in adult neutrophils. Strikingly, these neonate-specific cellular properties seemed to be associated with no neutrophil infiltration into the injured spinal cord, followed by significantly lower expression of inflammatory cytokines (Il-1β, Il-6 and TNF-α) after SCI in the spinal cords of neonates than in those of adults. At the same time, significantly fewer apoptotic neurons and greater axonal regeneration were observed in neonates in comparison with adults, which led to a marked recovery of locomotor function. This neonate-specific mechanism of inflammation regulation may have potential therapeutic applications in controlling inflammation after adult SCI.
Collapse
Affiliation(s)
- Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiichiro Iida
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Takeshi Maeda
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Osaka University, Osaka, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Li Y, Chen Y, Hu X, Ouyang F, Li J, Huang J, Ye J, Shan F, Luo Y, Yu S, Li Z, Yao F, Liu Y, Shi Y, Zheng M, Cheng L, Jing J. Fingolimod (FTY720) Hinders Interferon-γ-Mediated Fibrotic Scar Formation and Facilitates Neurological Recovery After Spinal Cord Injury. J Neurotrauma 2023; 40:2580-2595. [PMID: 36879472 DOI: 10.1089/neu.2022.0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Following spinal cord injury (SCI), fibrotic scar inhibits axon regeneration and impairs neurological function recovery. It has been reported that T cell-derived interferon (IFN)-γ plays a pivotal role in promoting fibrotic scarring in neurodegenerative disease. However, the role of IFN-γ in fibrotic scar formation after SCI has not been declared. In this study, a spinal cord crush injury mouse was established. Western blot and immunofluorescence showed that IFN-γ was surrounded by fibroblasts at 3, 7, 14, and 28 days post-injury. Moreover, IFN-γ is mainly secreted by T cells after SCI. Further, in situ injection of IFN-γ into the normal spinal cord resulted in fibrotic scar formation and inflammation response at 7 days post-injection. After SCI, the intraperitoneal injection of fingolimod (FTY720), a sphingosine-1-phosphate receptor 1 (S1PR1) modulator and W146, an S1PR1 antagonist, significantly reduced T cell infiltration, attenuating fibrotic scarring via inhibiting IFN-γ/IFN-γR pathway, while in situ injection of IFN-γ diminished the effect of FTY720 on reducing fibrotic scarring. FTY720 treatment inhibited inflammation, decreased lesion size, and promoted neuroprotection and neurological recovery after SCI. These findings demonstrate that the inhibition of T cell-derived IFN-γ by FTY720 suppressed fibrotic scarring and contributed to neurological recovery after SCI.
Collapse
Affiliation(s)
- Yiteng Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihao Chen
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xuyang Hu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangru Ouyang
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jianjian Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jinxin Huang
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jianan Ye
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangli Shan
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yong Luo
- Scientific Research and Experiment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuisheng Yu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziyu Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fei Yao
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanchang Liu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi Shi
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meige Zheng
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Cheng
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juehua Jing
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
15
|
Estera LA, Walsh SP, Headen JA, Williamson RE, Kalinski AL. Neuroinflammation: Breaking barriers and bridging gaps. Neurosci Res 2023; 197:9-17. [PMID: 34748905 DOI: 10.1016/j.neures.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/04/2023]
Abstract
Neurons are the cells of the nervous system and are responsible for every thought, movement and perception. Immune cells are the cells of the immune system, constantly protecting from foreign pathogens. Understanding the interaction between the two systems is especially important in disease states such as autoimmune or neurodegenerative disease. Unfortunately, this interaction is typically detrimental to the host. However, recent efforts have focused on how neurons and immune cells interact, either directly or indirectly, following traumatic injury to the nervous system. The outcome of this interaction can be beneficial - leading to successful neural repair, or detrimental - leading to functional deficits, depending on where the injury occurs. This review will discuss our understanding of neuron-immune cell interactions after traumatic injury to both the peripheral and central nervous system.
Collapse
Affiliation(s)
- Lora A Estera
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Sam P Walsh
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Jordan A Headen
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | | | - Ashley L Kalinski
- Department of Biology, Ball State University, Muncie, IN 47306, USA.
| |
Collapse
|
16
|
Kijima K, Ono G, Kobayakawa K, Saiwai H, Hara M, Yoshizaki S, Yokota K, Saito T, Tamaru T, Iura H, Haruta Y, Kitade K, Utsunomiya T, Konno D, Edgerton VR, Liu CY, Sakai H, Maeda T, Kawaguchi K, Matsumoto Y, Okada S, Nakashima Y. Zinc deficiency impairs axonal regeneration and functional recovery after spinal cord injury by modulating macrophage polarization via NF-κB pathway. Front Immunol 2023; 14:1290100. [PMID: 38022538 PMCID: PMC10666775 DOI: 10.3389/fimmu.2023.1290100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Background Spinal cord injury (SCI) is a devastating disease that results in permanent paralysis. Currently, there is no effective treatment for SCI, and it is important to identify factors that can provide therapeutic intervention during the course of the disease. Zinc, an essential trace element, has attracted attention as a regulator of inflammatory responses. In this study, we investigated the effect of zinc status on the SCI pathology and whether or not zinc could be a potential therapeutic target. Methods We created experimental mouse models with three different serum zinc concentration by changing the zinc content of the diet. After inducing contusion injury to the spinal cord of three mouse models, we assessed inflammation, apoptosis, demyelination, axonal regeneration, and the number of nuclear translocations of NF-κB in macrophages by using qPCR and immunostaining. In addition, macrophages in the injured spinal cord of these mouse models were isolated by flow cytometry, and their intracellular zinc concentration level and gene expression were examined. Functional recovery was assessed using the open field motor score, a foot print analysis, and a grid walk test. Statistical analysis was performed using Wilcoxon rank-sum test and ANOVA with the Tukey-Kramer test. Results In macrophages after SCI, zinc deficiency promoted nuclear translocation of NF-κB, polarization to pro-inflammatory like phenotype and expression of pro-inflammatory cytokines. The inflammatory response exacerbated by zinc deficiency led to worsening motor function by inducing more apoptosis of oligodendrocytes and demyelination and inhibiting axonal regeneration in the lesion site compared to the normal zinc condition. Furthermore, zinc supplementation after SCI attenuated these zinc-deficiency-induced series of responses and improved motor function. Conclusion We demonstrated that zinc affected axonal regeneration and motor functional recovery after SCI by negatively regulating NF-κB activity and the subsequent inflammatory response in macrophages. Our findings suggest that zinc supplementation after SCI may be a novel therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeyuki Saito
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Utsunomiya
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - V. Reggie Edgerton
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
- Institut Guttmann. Hospital de Neurorehabilitació, Institut Universitari adscrit a la Universitat Autònoma de Barcelona, Barcelona, Badalona, Spain
| | - Charles Y. Liu
- Neurorestoration Center, University of Southern California, Los Angeles, CA, United States
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Rancho Research Institute, Los Amigos National Rehabilitation Center, Downey, CA, United States
| | - Hiroaki Sakai
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Fukushima Medical University, Fukushima, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
17
|
Wu X, Liu J, Li W, Khan MF, Dai H, Tian J, Priya R, Tian DJ, Wu W, Yaacoub A, Gu J, Syed F, Yu CH, Gao X, Yu Q, Xu XM, Brutkiewicz RR. CD1d-dependent neuroinflammation impairs tissue repair and functional recovery following a spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562047. [PMID: 37905092 PMCID: PMC10614755 DOI: 10.1101/2023.10.13.562047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Tissue damage resulting from a spinal cord injury (SCI) is primarily driven by a robust neuroimmune/neuroinflammatory response. This intricate process is mainly governed by a multitude of cytokines and cell surface proteins in the central nervous system (CNS). However, the critical components of the neuroimmune/neuroinflammatory response during SCI are still not well-defined. In this study, we investigated the impact of CD1d, an MHC class I-like molecule mostly known for presenting lipid antigens to natural killer T (NKT) cells and regulating immune/inflammatory responses, on neuroimmune/neuroinflammatory responses induced by SCI. We observed an increased expression of CD1d on various cell types within the spinal cord, including microglia/macrophages, oligodendrocytes (ODCs), and endothelial cells (DCs), but not on neurons or astrocytes post-SCI. In comparison to wildtype (WT) mice, a T10 contusive SCI in CD1d knockout (CD1dKO or Cd1d -/- ) mice resulted in markedly reduced proinflammatory cytokine release, microglia/macrophage activation and proliferation. Following SCI, the levels of inflammatory cytokines and activation/proliferation of microglia/macrophages were dramatically reduced, while anti-inflammatory cytokines such as IL-4 and growth factors like VEGF were substantially increased in the spinal cord tissues of CD1dKO mice when compared to WT mice. In the post-acute phase of SCI (day 7 post-SCI), CD1dKO mice had a significantly higher frequency of tissue-repairing macrophages, but not other types of immune cells, in the injured spinal cord tissues compared to WT mice. Moreover, CD1d-deficiency protected spinal cord neuronal cells and tissue, promoting functional recovery after a SCI. However, the neuroinflammation in WT mouse spinal cords was independent of the canonical CD1d/NKT cell axis. Finally, treatment of injured mice with a CD1d-specific monoclonal antibody significantly enhanced neuroprotection and improved functional recovery. Therefore, CD1d promotes the proinflammatory response following a SCI and represents a potential therapeutic target for spinal cord repair. Significance Statement The cell surface molecule, CD1d, is known to be recognized by cells of the immune system. To our knowledge, this is the first observation that the CD1d molecule significantly contributes to neuroinflammation following a spinal cord injury (SCI) in a manner independent of the CD1d/NKT cell axis. This is important, because this work reveals CD1d as a potential therapeutic target following an acute SCI for which there are currently no effective treatments.
Collapse
|
18
|
Ono G, Kobayakawa K, Saiwai H, Tamaru T, Iura H, Haruta Y, Kitade K, Iida K, Kawaguchi K, Matsumoto Y, Tsuda M, Tamura T, Ozato K, Inoue K, Konno DJ, Maeda T, Okada S, Nakashima Y. Macrophages play a leading role in determining the direction of astrocytic migration in spinal cord injury via ADP-P2Y1R axis. Sci Rep 2023; 13:11177. [PMID: 37429920 DOI: 10.1038/s41598-023-38301-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
After spinal cord injury (SCI), inflammatory cells such as macrophages infiltrate the injured area, and astrocytes migrate, forming a glial scar around macrophages. The glial scar inhibits axonal regeneration, resulting in significant permanent disability. However, the mechanism through which glial scar-forming astrocytes migrate to the injury site has not been clarified. Here we show that migrating macrophages attract reactive astrocytes toward the center of the lesion after SCI. Chimeric mice with bone marrow lacking IRF8, which controls macrophage centripetal migration after SCI, showed widely scattered macrophages in the injured spinal cord with the formation of a huge glial scar around the macrophages. To determine whether astrocytes or macrophages play a leading role in determining the directions of migration, we generated chimeric mice with reactive astrocyte-specific Socs3-/- mice, which showed enhanced astrocyte migration, and bone marrow from IRF8-/- mice. In this mouse model, macrophages were widely scattered, and a huge glial scar was formed around the macrophages as in wild-type mice that were transplanted with IRF8-/- bone marrow. In addition, we revealed that macrophage-secreted ATP-derived ADP attracts astrocytes via the P2Y1 receptor. Our findings revealed a mechanism through which migrating macrophages attract astrocytes and affect the pathophysiology and outcome after SCI.
Collapse
Affiliation(s)
- Gentaro Ono
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Hirokazu Saiwai
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Tamaru
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hirotaka Iura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yohei Haruta
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuki Kitade
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keiichiro Iida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Kawaguchi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0395, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, Section on Molecular Genetics of Immunity, Division of Developmental Biology, NICHD, National Institutes of Health, Building 6A, Room 2A01, 6 Center Drive, Bethesda, MD, 20892, USA
| | - Kazuhide Inoue
- Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka-shi, Fukuoka, 819-0395, Japan
- Greenpharma Research Center for System Drug Discovery, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Dai-Jiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, 577-8502, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, 550-4 Igisu, Iizuka, Fukuoka, 820-8508, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
19
|
Wu C, Shi L, Ma Y, Pan Y, Wang L, Chen S, Zhang Y, Wang J, Liu M, Guo Y. Construction and optimization of a coculture system of mouse brain microvascular endothelial cells and myelin debris. Neurosci Lett 2023:137345. [PMID: 37308055 DOI: 10.1016/j.neulet.2023.137345] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
Microvascular endothelial cells are a newly discovered cell type involved in the phagocytosis of myelin debris, which play a key role in the repair of spinal cord injuries. Several methods for the preparation of myelin debris and parameters for constructing a coculture system of microvascular endothelial cells and myelin debris are available, but no systematic studies have yet been conducted, which hinders further exploration of the mechanisms of demyelinating disease repair. Herein, we aimed to develop a standardized method for this process. Myelin debris of different sizes was obtained from the brains of C57BL/6 mice by stripping the brains under aseptic conditions, multiple grinding, gradient centrifugation, etc. Transmission electron microscopy and nanoparticle size analysis were used to characterize myelin debris. Microvascular endothelial cells were cultured on a matrix gel, and myelin debris of different sizes (fluorescently labeled using CFSE) was placed in coculture after forming a vascular-like structure. Subsequently, myelin debris of different concentrations was cocultured in the vascular-like structure, and phagocytosis of myelin debris by microvascular endothelial cells was detected using immunofluorescence staining and flow cytometry. We found that myelin debris could be successfuly obtained from the mouse brain with secondary grinding and other steps and cocultured with microvascular endothelial cells at a concentration of 2 mg/mL, which promoted the phagocytosis of microvascular endothelial cells. In conclusion, we provide a reference for the protocol of a coculture system of microvascular endothelial cells and myelin debris.
Collapse
Affiliation(s)
- Chengjie Wu
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Shi
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong Ma
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yalan Pan
- Laboratory of Chinese Medicine Nursing Intervention for Chronic Diseases, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lining Wang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sixian Chen
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Mengmin Liu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yang Guo
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China.
| |
Collapse
|
20
|
Shafqat A, Albalkhi I, Magableh HM, Saleh T, Alkattan K, Yaqinuddin A. Tackling the glial scar in spinal cord regeneration: new discoveries and future directions. Front Cell Neurosci 2023; 17:1180825. [PMID: 37293626 PMCID: PMC10244598 DOI: 10.3389/fncel.2023.1180825] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Axonal regeneration and functional recovery are poor after spinal cord injury (SCI), typified by the formation of an injury scar. While this scar was traditionally believed to be primarily responsible for axonal regeneration failure, current knowledge takes a more holistic approach that considers the intrinsic growth capacity of axons. Targeting the SCI scar has also not reproducibly yielded nearly the same efficacy in animal models compared to these neuron-directed approaches. These results suggest that the major reason behind central nervous system (CNS) regeneration failure is not the injury scar but a failure to stimulate axon growth adequately. These findings raise questions about whether targeting neuroinflammation and glial scarring still constitute viable translational avenues. We provide a comprehensive review of the dual role of neuroinflammation and scarring after SCI and how future research can produce therapeutic strategies targeting the hurdles to axonal regeneration posed by these processes without compromising neuroprotection.
Collapse
|
21
|
Zheng B, He Y, Yin S, Zhu X, Zhao Q, Yang H, Wang Z, Zhu R, Cheng L. Unresolved Excess Accumulation of Myelin-Derived Cholesterol Contributes to Scar Formation after Spinal Cord Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0135. [PMID: 37223476 PMCID: PMC10202378 DOI: 10.34133/research.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/25/2023]
Abstract
Spinal cord injury triggers complex pathological cascades, resulting in destructive tissue damage and incomplete tissue repair. Scar formation is generally considered a barrier for regeneration in the central nervous system. However, the intrinsic mechanism of scar formation after spinal cord injury has not been fully elucidated. Here, we report that excess cholesterol accumulates in phagocytes and is inefficiently removed from spinal cord lesions in young adult mice. Interestingly, we observed that excessive cholesterol also accumulates in injured peripheral nerves but is subsequently removed by reverse cholesterol transport. Meanwhile, preventing reverse cholesterol transport leads to macrophage accumulation and fibrosis in injured peripheral nerves. Furthermore, the neonatal mouse spinal cord lesions are devoid of myelin-derived lipids and can heal without excess cholesterol accumulation. We found that transplantation of myelin into neonatal lesions disrupts healing with excessive cholesterol accumulation, persistent macrophage activation, and fibrosis. Myelin internalization suppresses macrophage apoptosis mediated by CD5L expression, indicating that myelin-derived cholesterol plays a critical role in impaired wound healing. Taken together, our data suggest that the central nervous system lacks an efficient approach for cholesterol clearance, resulting in excessive accumulation of myelin-derived cholesterol, thereby inducing scar formation after injury.
Collapse
Affiliation(s)
- Bolin Zheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Yijing He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Shuai Yin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Xu Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Qing Zhao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Clinical Center for Brain and Spinal Cord Research,
Tongji University, Shanghai 200092, China
| |
Collapse
|
22
|
Yao F, Luo Y, Chen Y, Li Y, Hu X, You X, Li Z, Yu S, Tian D, Zheng M, Cheng L, Jing J. Myelin Debris Impairs Tight Junctions and Promotes the Migration of Microvascular Endothelial Cells in the Injured Spinal Cord. Cell Mol Neurobiol 2023; 43:741-756. [PMID: 35147836 DOI: 10.1007/s10571-022-01203-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/31/2022] [Indexed: 01/08/2023]
Abstract
Clearance of myelin debris caused by acute demyelination is an essential process for functional restoration following spinal cord injury (SCI). Microvascular endothelial cells, acting as "amateur" phagocytes, have been confirmed to engulf and degrade myelin debris, promoting the inflammatory response, robust angiogenesis, and persistent fibrosis. However, the effect of myelin debris engulfment on the function of endothelial tight junctions (TJs) remains unclear. Here, we demonstrate that myelin debris uptake impairs TJs and gap junctions of endothelial cells in the lesion core of the injured spinal cord and in vitro, resulting in increased permeability and leakage. We further show that myelin debris acts as an inducer to regulate the endothelial-to-mesenchymal transition in a dose-dependent manner and promotes endothelial cell migration through the PI3K/AKT and ERK signaling pathways. Together, our results indicate that myelin debris engulfment impairs TJs and promotes the migration of endothelial cells. Accelerating myelin debris clearance may help maintain blood-spinal cord barrier integrity, thus facilitating restoration of motor and sensory function following SCI.
Collapse
Affiliation(s)
- Fei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Yiteng Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Xingyu You
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Ziyu Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Shuisheng Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Dasheng Tian
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
23
|
Gu G, Zhu B, Ren J, Song X, Fan B, Ding H, Shang J, Wu H, Li J, Wang H, Li J, Wei Z, Feng S. Ang-(1-7)/MasR axis promotes functional recovery after spinal cord injury by regulating microglia/macrophage polarization. Cell Biosci 2023; 13:23. [PMID: 36739421 PMCID: PMC9899400 DOI: 10.1186/s13578-023-00967-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/19/2023] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory response is an essential part of secondary injury after spinal cord injury (SCI). During this period, the injury may be exacerbated through the release of a large number of inflammatory factors and the polarization of infiltrating macrophages and microglia towards M1. Ang-(1-7), mainly generated by Ang II via angiotensin-converting enzyme 2 (ACE2), can specifically bind to the G protein-coupled receptor Mas (MasR) and plays an important role in regulating inflammation and alleviating oxidative stress. METHODS We aimed to investigate whether activating the Ang-(1-7)/MasR axis in rats after SCI can regulate local neuroinflammation to achieve functional recovery and obtain its potential mechanism. MasR expression of bone marrow-derived macrophages was determined by Western blot. Immunofluorescence, Western blot, Flow cytometry, and RT-qPCR were applied to evaluate the polarization of Ang-(1-7) on macrophages and the regulation of inflammatory cytokines. Previous evaluation of the spinal cord and bladder after SCI was conducted by hematoxylin-eosin staining, Basso, Beattie, and Bresnahan (BBB) score, inclined plate test, electrophysiology, and catwalk were used to evaluate the functional recovery of rats. RESULTS MasR expression increased in macrophages under inflammatory conditions and further elevated after Ang-(1-7) treatment. Both in vivo and in vitro results confirmed that Ang-(1-7) could regulate the expression of inflammatory cytokines by down-regulating proinflammatory cytokines and up-regulating anti-inflammatory cytokines, and bias the polarization direction of microglia/macrophages to M2 phenotypic. After SCI, Ang-(1-7) administration in situ led to better histological and functional recovery in rats, and this recovery at least partly involved the TLR4/NF-κB signaling pathway. CONCLUSION As shown in our data, activating Ang-(1-7)/MasR axis can effectively improve the inflammatory microenvironment after spinal cord injury, promote the polarization of microglia/macrophages towards the M2 phenotype, and finally support the recovery of motor function. Therefore, we suggest using Ang-(1-7) as a feasible treatment strategy for spinal cord injury to minimize the negative consequences of the inflammatory microenvironment after spinal cord injury.
Collapse
Affiliation(s)
- Guangjin Gu
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Bin Zhu
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Jie Ren
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Xiaomeng Song
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Baoyou Fan
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Han Ding
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Jun Shang
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Heng Wu
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junjin Li
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Hongda Wang
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Jinze Li
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China
| | - Zhijian Wei
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China ,Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong China
| | - Shiqing Feng
- grid.412645.00000 0004 1757 9434Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, National Spinal Cord Injury International Cooperation Base, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin, 300052 China ,Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Shandong University, Jinan, Shandong China
| |
Collapse
|
24
|
Ono G, Kobayakawa K, Saiwai H, Tamaru T, lura H, Haruta Y, Kitade K, Iida KI, Kawaguchi KI, Matsumoto Y, Tsuda M, Tamura T, Ozato K, Inoue K, Konno DJ, Maeda T, Okada S, Nakashima Y. Macrophages play a leading role in determining the direction of astrocytic migration in spinal cord injury via ADP-P2Y1R axis. RESEARCH SQUARE 2023:rs.3.rs-2427082. [PMID: 36789440 PMCID: PMC9928047 DOI: 10.21203/rs.3.rs-2427082/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
After spinal cord injury (SCI), inflammatory cells such as macrophages infiltrate the injured area, and astrocytes migrate, forming a glial scar around macrophages. The glial scar inhibits axonal regeneration, resulting in significant permanent disability. However, the mechanism by which glial scar-forming astrocytes migrate to the injury site has not been clarified. Here we show that migrating macrophages attract reactive astrocytes toward the center of the lesion after SCI. Chimeric mice with bone marrow lacking IRF8, which controls macrophage centripetal migration after SCI, showed widely scattered macrophages in injured spinal cord with the formation of a huge glial scar around the macrophages. To determine whether astrocytes or macrophages play a leading role in determining the directions of migration, we generated chimeric mice with reactive astrocyte-specific Socs3 -/- mice, which showed enhanced astrocyte migration, and bone marrow from IRF8 -/- mice. In this mouse model, macrophages were widely scattered, and a huge glial scar was formed around the macrophages as in wild-type mice that were transplanted with IRF8 -/ bone marrow. In addition, we revealed that macrophage-secreted ATP-derived ADP attracts astrocytes via the P2Y1 receptor. Our findings revealed a mechanism in which migrating macrophages attracted astrocytes and affected the pathophysiology and outcome after SCI.
Collapse
Affiliation(s)
- Gentaro Ono
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hirokazu Saiwai
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Tamaru
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hirotaka lura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yohei Haruta
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazuki Kitade
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kei-Ichiro Iida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Kawaguchi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka Nishi-ku Fukuoka-shi Fukuoka 819-0395, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama,236-0004, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, NICHD, National Institutes of Health, Section on Molecular Genetics of Immunity, Building 6A, Room 2A01, 6 Center Drive, Bethesda, MD 20892, USA
| | - Kazuhide Inoue
- Kyushu University Institute for Advanced Study, Kyushu University, 744 Motooka Nishi-ku Fukuoka-shi Fukuoka 819-0395, Japan; Greenpharma Research Center for System Drug Discovery, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Dai-Jiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka 577-8502, Japan
| | - Takeshi Maeda
- Department of Orthopaedic Surgery, Spinal Injuries Center, 550-4 Igisu, Iizuka, Fukuoka, 820-8508, Japan
| | - Seiji Okada
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, Suita 565-0871, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
25
|
Tamaru T, Kobayakawa K, Saiwai H, Konno D, Kijima K, Yoshizaki S, Hata K, Iura H, Ono G, Haruta Y, Kitade K, Iida KI, Kawaguchi KI, Matsumoto Y, Kubota K, Maeda T, Okada S, Nakashima Y. Glial scar survives until the chronic phase by recruiting scar-forming astrocytes after spinal cord injury. Exp Neurol 2023; 359:114264. [PMID: 36336030 DOI: 10.1016/j.expneurol.2022.114264] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Spinal cord injury (SCI) causes reactive astrogliosis, the sequential phenotypic change of astrocytes in which naïve astrocytes (NAs) transform into reactive astrocytes (RAs) and subsequently become scar-forming astrocytes (SAs), resulting in glial scar formation around the lesion site and thereby limiting axonal regeneration and motor/sensory functional recovery. Inhibiting the transformation of RAs into SAs in the acute phase attenuates the reactive astrogliosis and promotes regeneration. However, whether or not SAs once formed can revert to RAs or SAs is unclear. We performed selective isolation of astrocytes from glial scars at different time points for a gene expression analysis and found that the expression of Sox9, an important transcriptional factor for glial cell differentiation, was significantly increased in chronic phase astrocytes (CAs) compared to SAs in the sub-acute phase. Furthermore, CAs showed a significantly lower expression of chondroitin sulfate proteoglycan (CSPG)-related genes than SAs. These results indicated that SAs changed their phenotypes according to the surrounding environment of the injured spinal cord over time. Even though the integrin-N-cadherin pathway is critical for glial scar formation, collagen-I-grown scar-forming astrocytes (Col-I-SAs) did not change their phenotype after depleting the effect of integrin or N-cadherin. In addition, we found that Col-I-SAs transplanted into a naïve spinal cord formed glial scar again by maintaining a high expression of genes involved in the integrin-N-cadherin pathway and a low expression of CSPG-related genes. Interestingly, the transplanted Col-I-SAs changed NAs into SAs, and anti-β1-integrin antibody blocked the recruitment of SAs while reducing the volume of glial scar in the chronic phase. Our findings indicate that while the characteristics of glial scars change over time after SCI, SAs have a cell-autonomous function to form and maintain a glial scar, highlighting the basic mechanism underlying the persistence of glial scars after central nervous system injury until the chronic phase, which may be a therapeutic target.
Collapse
Affiliation(s)
- Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering Kindai University, Higashiosaka, Japan
| | - Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Orthopedic Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Kazuhiro Hata
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei-Ichiro Iida
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-Ichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Kubota
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takeshi Maeda
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Osaka University, Suita, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
26
|
Fu SP, Chen SY, Pang QM, Zhang M, Wu XC, Wan X, Wan WH, Ao J, Zhang T. Advances in the research of the role of macrophage/microglia polarization-mediated inflammatory response in spinal cord injury. Front Immunol 2022; 13:1014013. [PMID: 36532022 PMCID: PMC9751019 DOI: 10.3389/fimmu.2022.1014013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022] Open
Abstract
It is often difficult to regain neurological function following spinal cord injury (SCI). Neuroinflammation is thought to be responsible for this failure. Regulating the inflammatory response post-SCI may contribute to the recovery of neurological function. Over the past few decades, studies have found that macrophages/microglia are one of the primary effector cells in the inflammatory response following SCI. Growing evidence has documented that macrophages/microglia are plastic cells that can polarize in response to microenvironmental signals into M1 and M2 macrophages/microglia. M1 produces pro-inflammatory cytokines to induce inflammation and worsen tissue damage, while M2 has anti-inflammatory activities in wound healing and tissue regeneration. Recent studies have indicated that the transition from the M1 to the M2 phenotype of macrophage/microglia supports the regression of inflammation and tissue repair. Here, we will review the role of the inflammatory response and macrophages/microglia in SCI and repair. In addition, we will discuss potential molecular mechanisms that induce macrophage/microglia polarization, with emphasis on neuroprotective therapies that modulate macrophage/microglia polarization, which will provide new insights into therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qi-Ming Pang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiang-Chong Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xue Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei-Hong Wan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,Collaborative Innovation Center of Chinese Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China,The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China,*Correspondence: Tao Zhang,
| |
Collapse
|
27
|
Deficiency of CD93 exacerbates inflammation-induced activation and migration of BV2 microglia by regulating the TAK1/NF-κB pathway. Neurosci Lett 2022; 791:136914. [DOI: 10.1016/j.neulet.2022.136914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/26/2022] [Accepted: 10/10/2022] [Indexed: 11/18/2022]
|
28
|
Li J, Liu X, Wu H, Guo P, Li B, Wang J, Tian W, Chen D, Gao M, Zhou Z, Liu S. Identification of hub genes related to the innate immune response activated during spinal cord injury. FEBS Open Bio 2022; 12:1839-1856. [PMID: 36047918 PMCID: PMC9527585 DOI: 10.1002/2211-5463.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/01/2022] [Accepted: 08/17/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) often leads to sensory and motor dysfunction. Two major factors that hinder spinal cord repair are local inflammation and glial scar formation after SCI, and thus appropriate immunotherapy may alleviate damage. To characterize changes in gene expression that occur during SCI and thereby identify putative targets for immunotherapy, here we analyzed the dataset GSE5296 (containing one control group and six SCI groups at different timepoints) to identify differentially-expressed genes. Functional enrichment analysis was performed and a protein-protein interaction network was created to identify possible hub genes. Finally, we performed quantitative PCR to verify changes in gene expression. The CIBERSORT algorithm was used to analyze innate immune cell infiltration patterns. The dataset GSE162610 (containing one control group and three SCI groups at different timepoints) was analyzed to evaluate innate immune cell infiltration at the single-cell level. The dataset GSE151371 (containing one control group [n = 10] and an SCI group [n = 38]) was used to detect the expression of hub genes in the blood from SCI patients. Differentially-expressed innate immune-related genes at each timepoint were identified, and the functions and related signaling pathways of these genes were examined. Six hub genes were identified and verified. We then analyzed the expression characteristics of these hub genes and characteristics of innate immune infiltration in SCI; finally, we examined ligand expression in the context of the CCL signaling pathway and COMPLEMENT signaling pathway networks. This study reveals the characteristics of innate immune cell infiltration and temporal expression patterns of hub genes, and may aid in the development of immunotherapies for SCI.
Collapse
Affiliation(s)
- Jianfeng Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Huachuan Wu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Baoliang Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Jianmin Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Wei Tian
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopedics and TraumatologyBeijing Jishuitan HospitalChina
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopedics and TraumatologyBeijing Jishuitan HospitalChina
| | - Manman Gao
- Department of Sport Medicine, Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalChina,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical EngineeringShenzhen University Health Science CenterChina
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Shaoyu Liu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
29
|
Corrigendum: Purinergic signaling systems across comparative models of spinal cord injury. Neural Regen Res 2022; 18:689-696. [PMID: 36018196 PMCID: PMC9727416 DOI: 10.4103/1673-5374.350234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
[This corrects the article DOI: 10.4103/1673-5374.338993].
Collapse
|
30
|
Fan C, Yang W, Zhang L, Cai H, Zhuang Y, Chen Y, Zhao Y, Dai J. Restoration of spinal cord biophysical microenvironment for enhancing tissue repair by injury-responsive smart hydrogel. Biomaterials 2022; 288:121689. [DOI: 10.1016/j.biomaterials.2022.121689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 11/02/2022]
|
31
|
Circular RNA Hecw1 Regulates the Inflammatory Imbalance in Spinal Cord Injury via miR-3551-3p/LRRTM1 Axis. Appl Biochem Biotechnol 2022; 194:5151-5166. [PMID: 35699802 DOI: 10.1007/s12010-022-03999-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/02/2022]
Abstract
Spinal cord injury (SCI) is a neurological disease having devastating effect and results in the development of systemic inflammation. However, the molecular mechanisms of SCI remain not entirely elucidated. This study was directed toward exploring the circ Hecw1 involved in the mechanism of lipopolysaccharide (LPS)-triggered inflammation damage in neuronal cells. The in vitro model of SCI based on PC12 cells were established with lipopolysaccharide. The cell proliferation was determined by the use of cell counting kit-8 (CCK8). The expressions of circHecw1, miR-3551-3p, and inflammatory factors were measured by quantitative real-time PCR and ELISA assay. Flow cytometry was used to assess apoptosis. Western blot analysis was performed for the purpose of determining LRRTM1 and NF-kB signaling. The expression of circ Hecw1, TNF-α, IL-6, and IL-1β in LPS-triggered PC12 cells and the expression of miR-3551-3p and IL-10 were significantly decreased. Knockdown of circHecw1 promoted proliferation and inhibited apoptosis and reduction in the inflammatory cytokine expression. Our study revealed that circHecw1 regulates SCI neuronal cell inflammation imbalance by regulating the miR-3551-3p/LRRTM1 signaling.
Collapse
|
32
|
An anti-inflammatory and neuroprotective biomimetic nanoplatform for repairing spinal cord injury. Bioact Mater 2022; 18:569-582. [PMID: 35845318 PMCID: PMC9256979 DOI: 10.1016/j.bioactmat.2022.05.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/23/2022] Open
Abstract
Spinal cord regeneration after a spinal cord injury (SCI) remains a difficult challenge due to the complicated inflammatory microenvironment and neuronal damage at the injury sites. In this study, retinoic acid (RA) and curcumin (Cur) were co-loaded with bovine serum albumin (BSA) self-assembly to obtain RA@BSA@Cur nanoparticles (NPs) for SCI treatment. Cur, as an antioxidant drug, facilitated reactive oxygen species (ROS) scavenging, and decreased the amount of inflammatory factors secreted by macrophages, while RA could enhance neurite extensions and neural differentiation. The constructed RA@BSA@Cur NPs not only induced polarization of macrophages toward pro-regenerative phenotypes and markedly reduced the inflammatory response of macrophages or microglia, but also increased neurite length in PC12 cells and neuronal differentiation of bone marrow mesenchymal stem cells, improved the differentiation of neural stem cells (NSCs) into β3-tubulin+ neurons, and reversed the pro-astrocyte differentiation effect of inflammatory cytokines on NSCs. In vivo experiments revealed that RA@BSA@Cur NPs regulated the phenotypic polarization of macrophages, inhibited the release of inflammatory mediators, promoted functional neuron regeneration and motor function, and further inhibited scar tissue formation. This study highlighted that the BSA-based biomimetic nanomaterials could be used as ROS scavengers and nerve regeneration promoters for treating SCI. Retinoic acid and Curcumin with remarkable neuroprotective and antioxidant activities are prepared for spinal cord injury. RA@BSA@Cur NPs induce polarization of macrophages, reduce levels of tumor necrosis factor-α and interleukin-6, and thus reduce the inflammatory response. RA@BSA@Cur NPs increase neurite length in PC12 cells and neuronal differentiation of bone marrow mesenchymal stem cells, and regulate the neurons/astrocytes differentiation of NSCs. RA@BSA@Cur NPs reverse the pro-astrocyte differentiation effect of macrophages-derived inflammatory cytokines on NSCs. RA@BSA@Cur NPs promote neuron regeneration and motor function, inhibit scar tissue formation, and regulate the phenotypic polarization of macrophages.
Collapse
|
33
|
Herr SA, Gardeen SS, Low PS, Shi R. Targeted delivery of acrolein scavenger hydralazine in spinal cord injury using folate-linker-drug conjugation. Free Radic Biol Med 2022; 184:66-73. [PMID: 35398493 DOI: 10.1016/j.freeradbiomed.2022.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 11/26/2022]
Abstract
Oxidative stress has been shown to play a critical pathogenic role in functional loss after spinal cord injury (SCI). As a direct result of oxidative stress, lipid peroxidation-derived aldehydes have emerged as key culprits that sustain secondary injury and contribute significantly to pathological outcomes. Acrolein, a neurotoxin, has been shown to be elevated in SCI and can result in post-SCI neurological deficits. Reducing acrolein has therefore emerged as a novel and effective therapeutic strategy in SCI. Previous studies have revealed that hydralazine, an FDA approved blood pressure lowering medication, when administered after SCI shows strong acrolein scavenging capabilities and significantly improves cellular and behavioral outcomes. However, while effective at scavenging acrolein, hydralazine's blood pressure lowering activity can have a detrimental impact on neurotrauma patients. Here, our goal was to preserve the acrolein scavenging capability while mitigating the effect of hydralazine on blood pressure. We accomplished this using a folate-targeted delivery system to deploy hydralazine to the folate receptor positive inflammatory site of the cord injury. Using a model of rat SCI, we found that this system is effective for targeting the injury site, and that folate targeted hydralazine can scavenge acrolein without significantly impacting blood pressure.
Collapse
Affiliation(s)
- Seth A Herr
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| | - Spencer S Gardeen
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Philip S Low
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| |
Collapse
|
34
|
Liu W, Zhao Y, Wang G, Feng S, Ge X, Ye W, Wang Z, Zhu Y, Cai W, Bai J, Zhou X. TRIM22 inhibits osteosarcoma progression through destabilizing NRF2 and thus activation of ROS/AMPK/mTOR/autophagy signaling. Redox Biol 2022; 53:102344. [PMID: 35636015 PMCID: PMC9144049 DOI: 10.1016/j.redox.2022.102344] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/11/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is a malignant bone tumor that mainly occurs in adolescents. It is accompanied by a high rate of lung metastasis, and high mortality. Recent studies have suggested the important roles of tripartite motif-containing (TRIM) family proteins in regulating various substrates and signaling pathways in different tumors. However, the detailed functional role of TRIM family proteins in the progression of OS is still unknown and requires further investigations. In this study, we found that tripartite motif-containing 22 (TRIM22) was downregulated in OS tissues and was hence associated with better prognosis. In vitro and in vivo functional analysis demonstrated that TRIM22 inhibits proliferation and metastasis of OS cells. Nuclear factor erythroid 2-related factor 2 (NRF2), a redox regulator, was identified as a novel target for TRIM22. TRIM22 interacts with and accelerates the degradation of NRF2 by inducing its ubiquitination dependent on its E3 ligase activity but independent of Kelch-like ECH-associated protein 1 (KEAP1). Further, a series of gain- and loss-of-function experiments showed that knockdown or overexpression of NRF2 reversed the functions of knockdown or overexpression of TRIM22 in OS. Mechanistically, TRIM22 inhibited OS progression through NRF2-mediated intracellular reactive oxygen species (ROS) imbalance. ROS production was significantly promoted and mitochondrial potential was remarkably inhibited when overexpressing TRIM22, thus activating AMPK/mTOR signaling. Moreover, TRIM22 was also found to inhibit Warburg effect in OS cells. Autophagy activation was found in OS cells which were overexpressed TRIM22, thus leading to autophagic cell death. Treatment with N-Acetylcysteine (NAC), a ROS scavenger or the autophagy inhibitor 3-Methyladenine (3-MA) abolished the decreased malignant phenotypes in TRIM22 overexpressing OS cells. In conclusion, our study indicated that TRIM22 inhibits OS progression by promoting proteasomal degradation of NRF2 independent of KEAP1, thereby activating ROS/AMPK/mTOR/Autophagy signaling that leads to autophagic cell death in OS. Therefore, our findings indicated that targeting TRIM22/NRF2 could be a promising therapeutic target for treating OS. TRIM22 inhibits proliferation and metastasis of OS cells. TRIM22 interacts with and accelerates NRF2 degradation by inducing its ubiquitination dependent on E3 ligase activity. TRIM22 inhibited OS progression through NRF2-mediated intracellular ROS imbalance. TRIM22 inhibits OS progression by promoting NRF2 degradation, thereby activating ROS/AMPK/mTOR/Autophagy signaling.
Collapse
|
35
|
Li Z, Yu S, Liu Y, Hu X, Li Y, Xiao Z, Chen Y, Tian D, Xu X, Cheng L, Zheng M, Jing J. SU16f inhibits fibrotic scar formation and facilitates axon regeneration and locomotor function recovery after spinal cord injury by blocking the PDGFRβ pathway. J Neuroinflammation 2022; 19:95. [PMID: 35429978 PMCID: PMC9013464 DOI: 10.1186/s12974-022-02449-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 03/28/2022] [Indexed: 11/19/2022] Open
Abstract
Background Excessively deposited fibrotic scar after spinal cord injury (SCI) inhibits axon regeneration. It has been reported that platelet-derived growth factor receptor beta (PDGFRβ), as a marker of fibrotic scar-forming fibroblasts, can only be activated by platelet-derived growth factor (PDGF) B or PDGFD. However, whether the activation of the PDGFRβ pathway can mediate fibrotic scar formation after SCI remains unclear. Methods A spinal cord compression injury mouse model was used. In situ injection of exogenous PDGFB or PDGFD in the spinal cord was used to specifically activate the PDGFRβ pathway in the uninjured spinal cord, while intrathecal injection of SU16f was used to specifically block the PDGFRβ pathway in the uninjured or injured spinal cord. Immunofluorescence staining was performed to explore the distributions and cell sources of PDGFB and PDGFD, and to evaluate astrocytic scar, fibrotic scar, inflammatory cells and axon regeneration after SCI. Basso Mouse Scale (BMS) and footprint analysis were performed to evaluate locomotor function recovery after SCI. Results We found that the expression of PDGFD and PDGFB increased successively after SCI, and PDGFB was mainly secreted by astrocytes, while PDGFD was mainly secreted by macrophages/microglia and fibroblasts. In addition, in situ injection of exogenous PDGFB or PDGFD can lead to fibrosis in the uninjured spinal cord, while this profibrotic effect could be specifically blocked by the PDGFRβ inhibitor SU16f. We then treated the mice after SCI with SU16f and found the reduction of fibrotic scar, the interruption of scar boundary and the inhibition of lesion and inflammation, which promoted axon regeneration and locomotor function recovery after SCI. Conclusions Our study demonstrates that activation of PDGFRβ pathway can directly induce fibrotic scar formation, and specific blocking of this pathway would contribute to the treatment of SCI.
Collapse
|
36
|
Jiang K, Sun Y, Chen X. Mechanism Underlying Acupuncture Therapy in Spinal Cord Injury: A Narrative Overview of Preclinical Studies. Front Pharmacol 2022; 13:875103. [PMID: 35462893 PMCID: PMC9021644 DOI: 10.3389/fphar.2022.875103] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/14/2022] [Indexed: 12/29/2022] Open
Abstract
Spinal cord injury (SCI) results from various pathogenic factors that destroy the normal structure and function of the spinal cord, subsequently causing sensory, motor, and autonomic nerve dysfunction. SCI is one of the most common causes of disability and death globally. It leads to severe physical and mental injury to patients and causes a substantial economic burden on families and the society. The pathological changes and underlying mechanisms within SCI involve oxidative stress, apoptosis, inflammation, etc. As a traditional therapy, acupuncture has a positive effect promoting the recovery of SCI. Acupuncture-induced neuroprotection includes several mechanisms such as reducing oxidative stress, inhibiting the inflammatory response and neuronal apoptosis, alleviating glial scar formation, promoting neural stem cell differentiation, and improving microcirculation within the injured area. Therefore, the recent studies exploring the mechanism of acupuncture therapy in SCI will help provide a theoretical basis for applying acupuncture and seeking a better treatment target and acupuncture approach for SCI patients.
Collapse
Affiliation(s)
- Kunpeng Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xinle Chen
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
- *Correspondence: Xinle Chen,
| |
Collapse
|
37
|
Qian ZY, Kong RY, Zhang S, Wang BY, Chang J, Cao J, Wu CQ, Huang ZY, Duan A, Li HJ, Yang L, Cao XJ. Ruxolitinib attenuates secondary injury after traumatic spinal cord injury. Neural Regen Res 2022; 17:2029-2035. [PMID: 35142693 PMCID: PMC8848590 DOI: 10.4103/1673-5374.335165] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Excessive inflammation post-traumatic spinal cord injury (SCI) induces microglial activation, which leads to prolonged neurological dysfunction. However, the mechanism underlying microglial activation-induced neuroinflammation remains poorly understood. Ruxolitinib (RUX), a selective inhibitor of JAK1/2, was recently reported to inhibit inflammatory storms caused by SARS-CoV-2 in the lung. However, its role in disrupting inflammation post-SCI has not been confirmed. In this study, microglia were treated with RUX for 24 hours and then activated with interferon-γ for 6 hours. The results showed that interferon-γ-induced phosphorylation of JAK and STAT in microglia was inhibited, and the mRNA expression levels of pro-inflammatory cytokines tumor necrosis factor-α, interleukin-1β, interleukin-6, and cell proliferation marker Ki67 were reduced. In further in vivo experiments, a mouse model of spinal cord injury was treated intragastrically with RUX for 3 successive days, and the findings suggest that RUX can inhibit microglial proliferation by inhibiting the interferon-γ/JAK/STAT pathway. Moreover, microglia treated with RUX centripetally migrated toward injured foci, remaining limited and compacted within the glial scar, which resulted in axon preservation and less demyelination. Moreover, the protein expression levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6 were reduced. The neuromotor function of SCI mice also recovered. These findings suggest that RUX can inhibit neuroinflammation through inhibiting the interferon-γ/JAK/STAT pathway, thereby reducing secondary injury after SCI and producing neuroprotective effects.
Collapse
Affiliation(s)
- Zhan-Yang Qian
- Spine Center, Zhongda Hospital of Southeast University; Medical School, Southeast University, Nanjing, Jiangsu Province, China
| | - Ren-Yi Kong
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sheng Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Bin-Yu Wang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Chang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiang Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chao-Qin Wu
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zi-Yan Huang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ao Duan
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hai-Jun Li
- Department of Orthopedics, Hospital Affiliated 5 to Nantong University (Taizhou People's Hospital); Taizhou Clinical Medical School of Nanjing Medical University, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Lei Yang
- Department of Orthopedics, Hospital Affiliated 5 to Nantong University (Taizhou People's Hospital); Taizhou Clinical Medical School of Nanjing Medical University, Taizhou People's Hospital, Taizhou, Jiangsu Province, China
| | - Xiao-Jian Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
38
|
Gao X, Cheng W, Zhang X, Zhou Z, Ding Z, Zhou X, Lu Q, Kaplan DL. Nerve Growth Factor-Laden Anisotropic Silk Nanofiber Hydrogels to Regulate Neuronal/Astroglial Differentiation for Scarless Spinal Cord Repair. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3701-3715. [PMID: 35006667 DOI: 10.1021/acsami.1c19229] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Scarless spinal cord regeneration remains a challenge due to the complicated microenvironment at lesion sites. In this study, the nerve growth factor (NGF) was immobilized in silk protein nanofiber hydrogels with hierarchical anisotropic microstructures to fabricate bioactive systems that provide multiple physical and biological cues to address spinal cord injury (SCI). The NGF maintained bioactivity inside the hydrogels and regulated the neuronal/astroglial differentiation of neural stem cells. The aligned microstructures facilitated the migration and orientation of cells, which further stimulated angiogenesis and neuron extensions both in vitro and in vivo. In a severe rat long-span hemisection SCI model, these hydrogel matrices reduced scar formation and achieved the scarless repair of the spinal cord and effective recovery of motor functions. Histological analysis confirmed the directional regenerated neuronal tissues, with a similar morphology to that of the normal spinal cord. The in vitro and in vivo results showed promising utility for these NGF-laden silk hydrogels for spinal cord regeneration while also demonstrating the feasibility of cell-free bioactive matrices with multiple cues to regulate endogenous cell responses.
Collapse
Affiliation(s)
- Xiang Gao
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China
| | - Weinan Cheng
- Department of Orthopedics, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, People's Republic of China
| | - Xiaoyi Zhang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhengyu Zhou
- Laboratory Animal Center, Medical Collagen of Soochow University, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215000, People's Republic of China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
39
|
Feng D, Yu J, Bao L, Fan D, Zhang B. Inhibiting RGS1 attenuates secondary inflammation response and tissue degradation via the TLR/TRIF/NF-κB pathway in macrophage post spinal cord injury. Neurosci Lett 2022; 768:136374. [PMID: 34852285 DOI: 10.1016/j.neulet.2021.136374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022]
Abstract
Macrophage-dominated inflammation by the activation of Toll-like receptor (TLR) pathway leads to neurological disruption after spinal cord injury (SCI). Regulator of G-protein signaling 1 (RGS1) is reported to be a regulator in inflammation. The present study thus purposes to identify the unknown role of RGS1 mediating TLR on inflammation post SCI. A mouse model of traumatic SCI was established by a mechanical trauma at T10. The mice underwent SCI and a macrophage line activated by lipopolysaccharide (LPS) were treated with shRNA-RGS1 to elucidate the role of RGS1 in inflammatory progression. The inflammatory factors were measured, and the degree of histology and function protection were determined. The expression levels of RGS1, myeloid differentiation primary response protein 88 (Myd88), (TIR-domain-containing adaptor inducing interferon-β (TRIF), p38, metalloproteinase (MMP)-2, and MMP-9 were determined. RGS1 was robustly increased both in LPS-activated macrophage and SCI mice. The TLR signaling pathway-induced inflammation was suppressed by RGS1 knockdown. shRNA-mediated silence of RGS1 was exhibited a prominent decrease in TNF-α, IL-1β and IL-6 via TLR/TRIF/ nuclear factor kappa-B (NF-κB) axis. Depletion of RGS1 also inhibited MMP-induced tissue degradation via MAPK-p38 pathway in SCI mice. Moreover, suppression of RGS1 improved spinal cord histology and function recovery. These findings suggest that RGS1 regulates inflammation and tissue disruption via TLR/TRIF/NF-κB signaling pathway in mice with SCI, thereby explaining a novel target that regulates macrophage inflammation post SCI.
Collapse
Affiliation(s)
- Dongqian Feng
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China; Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Jiasheng Yu
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Lei Bao
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Daobo Fan
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China
| | - Bin Zhang
- Department of Orthopedics, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Shuyang Hospital of Traditional Chinese Medicine, Shuyang 223600, China.
| |
Collapse
|
40
|
Luo Y, Yao F, Hu X, Li Y, Chen Y, Li Z, Zhu Z, Yu S, Tian D, Cheng L, Zheng M, Jing J. M1 macrophages impair tight junctions between endothelial cells after spinal cord injury. Brain Res Bull 2022; 180:59-72. [PMID: 34995751 DOI: 10.1016/j.brainresbull.2021.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/30/2021] [Accepted: 12/31/2021] [Indexed: 12/11/2022]
Abstract
After spinal cord injury (SCI), endogenous angiogenesis occurs in the injury core, unexpectedly accompanied by continuous leakage of the blood-spinal cord barrier (BSCB), which may be caused by destruction of the tight junctions (TJs) between vascular endothelial cells-an important structure of the BSCB. Blood-derived macrophages infiltrate into the spinal cord, aggregate to the injury core and then polarize toward M1/M2 phenotypes after SCI. However, the effect of macrophages with different polarizations on the TJs between vascular endothelial cells remains unclear. Here, we demonstrated that from 7 days postinjury (dpi) to 28 dpi, accompanied by the aggregation of macrophages, the expression of claudin-5 (CLN-5) and zonula occludens-1 (ZO-1) in vascular endothelial cells in the injury core was significantly decreased in comparison to that in normal spinal cord tissue and in the penumbra. Moreover, the leakage of the BSCB was severe in the injury core, as demonstrated by FITC-dextran perfusion. Notably, our study demonstrated that depletion of macrophages facilitated the restoration of TJs between vascular endothelial cells and decreased the leakage of BSCB in the injury core after SCI. Furthermore, we confirmed that the endothelial TJs could be impaired by M1 macrophages through secreting IL-6 in vitro, leading to an increased permeability of endothelial cells, but it was not significantly affected by M0 and M2 macrophages. These results indicated that the TJs between vascular endothelial cells were impaired by M1 macrophages in the injury core, potentially resulting in continuous leakage of the BSCB after SCI. Preventing M1 polarization of macrophages or blocking IL-6 in the injury core may promote restoration of the BSCB, thus accelerating functional recovery after SCI.
Collapse
Affiliation(s)
- Yang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Fei Yao
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Yiteng Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Yihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Ziyu Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Zhenyu Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Shuisheng Yu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Dasheng Tian
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China; School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China.
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China.
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, Anhui Province, People's Republic of China.
| |
Collapse
|
41
|
Ding Y, Zhang D, Wang S, Zhang X, Yang J. Hematogenous Macrophages: A New Therapeutic Target for Spinal Cord Injury. Front Cell Dev Biol 2021; 9:767888. [PMID: 34901013 PMCID: PMC8653770 DOI: 10.3389/fcell.2021.767888] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating disease leading to loss of sensory and motor functions, whose pathological process includes mechanical primary injury and secondary injury. Macrophages play an important role in SCI pathology. According to its origin, it can be divided into resident microglia and peripheral monocyte-derived macrophages (hematogenous Mφ). And it can also be divided into M1-type macrophages and M2-type macrophages on the basis of its functional characteristics. Hematogenous macrophages may contribute to the SCI process through infiltrating, scar forming, phagocytizing debris, and inducing inflammatory response. Although some of the activities of hematogenous macrophages are shown to be beneficial, the role of hematogenous macrophages in SCI remains controversial. In this review, following a brief introduction of hematogenous macrophages, we mainly focus on the function and the controversial role of hematogenous macrophages in SCI, and we propose that hematogenous macrophages may be a new therapeutic target for SCI.
Collapse
Affiliation(s)
- Yuanzhe Ding
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Di Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China
| | - Sheng Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
| | - Jingquan Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China
| |
Collapse
|
42
|
Jonavičė U, Romenskaja D, Kriaučiūnaitė K, Jarmalavičiūtė A, Pajarskienė J, Kašėta V, Tunaitis V, Malm T, Giniatulin R, Pivoriūnas A. Extracellular Vesicles from Human Teeth Stem Cells Trigger ATP Release and Promote Migration of Human Microglia through P2X4 Receptor/MFG-E8-Dependent Mechanisms. Int J Mol Sci 2021; 22:ijms222010970. [PMID: 34681627 PMCID: PMC8537493 DOI: 10.3390/ijms222010970] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) effectively suppress neuroinflammation and induce neuroprotective effects in different disease models. However, the mechanisms by which EVs regulate the neuroinflammatory response of microglia remains largely unexplored. Here, we addressed this issue by testing the action of EVs derived from human exfoliated deciduous teeth stem cells (SHEDs) on immortalized human microglial cells. We found that EVs induced a rapid increase in intracellular Ca2+ and promoted significant ATP release in microglial cells after 20 min of treatment. Boyden chamber assays revealed that EVs promoted microglial migration by 20%. Pharmacological inhibition of different subtypes of purinergic receptors demonstrated that EVs activated microglial migration preferentially through the P2X4 receptor (P2X4R) pathway. Proximity ligation and co-immunoprecipitation assays revealed that EVs promote association between milk fat globule-epidermal growth factor-factor VIII (MFG-E8) and P2X4R proteins. Furthermore, pharmacological inhibition of αVβ3/αVβ5 integrin suppressed EV-induced cell migration and formation of lipid rafts in microglia. These results demonstrate that EVs promote microglial motility through P2X4R/MFG-E8-dependent mechanisms. Our findings provide novel insights into the molecular mechanisms through which EVs target human microglia that may be exploited for the development of new therapeutic strategies targeting disease-associated neuroinflammation.
Collapse
Affiliation(s)
- Ugnė Jonavičė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Diana Romenskaja
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Karolina Kriaučiūnaitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Akvilė Jarmalavičiūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Justina Pajarskienė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Vytautas Kašėta
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Virginijus Tunaitis
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (T.M.); (R.G.)
| | - Rashid Giniatulin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland; (T.M.); (R.G.)
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; (U.J.); (D.R.); (K.K.); (A.J.); (J.P.); (V.K.); (V.T.)
- Correspondence:
| |
Collapse
|
43
|
Zhang P, Schlecht A, Wolf J, Boneva S, Laich Y, Koch J, Ludwig F, Boeck M, Thien A, Härdtner C, Kierdorf K, Agostini H, Schlunck G, Prinz M, Hilgendorf I, Wieghofer P, Lange C. The role of interferon regulatory factor 8 for retinal tissue homeostasis and development of choroidal neovascularisation. J Neuroinflammation 2021; 18:215. [PMID: 34544421 PMCID: PMC8454118 DOI: 10.1186/s12974-021-02230-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Microglia cells represent the resident innate immune cells of the retina and are important for retinal development and tissue homeostasis. However, dysfunctional microglia can have a negative impact on the structural and functional integrity of the retina under native and pathological conditions. METHODS In this study, we examined interferon-regulatory factor 8 (Irf8)-deficient mice to determine the transcriptional profile, morphology, and temporospatial distribution of microglia lacking Irf8 and to explore the effects on retinal development, tissue homeostasis, and formation of choroidal neovascularisation (CNV). RESULTS Our study shows that Irf8-deficient MG exhibit a considerable loss of microglial signature genes accompanied by a severely altered MG morphology. An in-depth characterisation by fundus photography, fluorescein angiography, optical coherence tomography and electroretinography revealed no major retinal abnormalities during steady state. However, in the laser-induced CNV model, Irf8-deficient microglia showed an increased activity of biological processes critical for inflammation and cell adhesion and a reduced MG cell density near the lesions, which was associated with significantly increased CNV lesion size. CONCLUSIONS Our results suggest that loss of Irf8 in microglia has negligible effects on retinal homeostasis in the steady state. However, under pathological conditions, Irf8 is crucial for the transformation of resident microglia into a reactive phenotype and thus for the suppression of retinal inflammation and CNV formation.
Collapse
Affiliation(s)
- Peipei Zhang
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anja Schlecht
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,Institute of Anatomy, Wuerzburg University, Wuerzburg, Germany
| | - Julian Wolf
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stefaniya Boneva
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Yannik Laich
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jana Koch
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Franziska Ludwig
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Myriam Boeck
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adrian Thien
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Carmen Härdtner
- Cardiology and Angiology, University Heart Center, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Medical Faculty, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg im Breisgau, Germany
| | - Hansjürgen Agostini
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Marco Prinz
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Faculty, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg im Breisgau, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany
| | - Ingo Hilgendorf
- Cardiology and Angiology, University Heart Center, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Peter Wieghofer
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany. .,Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Clemens Lange
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
44
|
Li Z, Wang Q, Hu H, Zheng W, Gao C. Research advances of biomaterials-based microenvironment-regulation therapies for repair and regeneration of spinal cord injury. Biomed Mater 2021; 16. [PMID: 34384071 DOI: 10.1088/1748-605x/ac1d3c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022]
Abstract
Traumatic spinal cord injury (SCI) usually results in restricted behaviour recovery and even life-changing paralysis, accompanied with numerous complications. Pathologically, the initial injuries trigger a series of secondary injuries, leading to an expansion of lesion site, a mass of neuron loss, and eventual failure of endogenous axon regeneration. As the advances rapidly spring up in regenerative medicine and tissue engineering biomaterials, regulation of these secondary injuries becomes possible, shedding a light on normal functional restoration. The successful tissue regeneration lies in proper regulation of the inflammatory microenvironment, including the inflammatory immune cells and inflammatory factors that lead to oxidative stress, inhibitory glial scar and neuroexcitatory toxicity. Specifically, the approaches based on microenvironment-regulating biomaterials have shown great promise in the repair and regeneration of SCI. In this review, the pathological inflammatory microenvironments of SCI are discussed, followed by the introduction of microenvironment-regulating biomaterials in terms of their impressive therapeutic effect in attenuation of secondary inflammation and promotion of axon regrowth. With the emphasis on regulating secondary events, the biomaterials for SCI treatment will become promising for clinical applications.
Collapse
Affiliation(s)
- Ziming Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China.,Dr Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
45
|
Li Z, Yu S, Hu X, Li Y, You X, Tian D, Cheng L, Zheng M, Jing J. Fibrotic Scar After Spinal Cord Injury: Crosstalk With Other Cells, Cellular Origin, Function, and Mechanism. Front Cell Neurosci 2021; 15:720938. [PMID: 34539350 PMCID: PMC8441597 DOI: 10.3389/fncel.2021.720938] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/28/2021] [Indexed: 01/18/2023] Open
Abstract
The failure of axonal regeneration after spinal cord injury (SCI) results in permanent loss of sensorimotor function. The persistent presence of scar tissue, mainly fibrotic scar and astrocytic scar, is a critical cause of axonal regeneration failure and is widely accepted as a treatment target for SCI. Astrocytic scar has been widely investigated, while fibrotic scar has received less attention. Here, we review recent advances in fibrotic scar formation and its crosstalk with other main cellular components in the injured core after SCI, as well as its cellular origin, function, and mechanism. This study is expected to provide an important basis and novel insights into fibrotic scar as a treatment target for SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Cheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Meige Zheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
46
|
Lu X, Lu F, Yu J, Xue X, Jiang H, Jiang L, Yang Y. Gramine promotes functional recovery after spinal cord injury via ameliorating microglia activation. J Cell Mol Med 2021; 25:7980-7992. [PMID: 34382745 PMCID: PMC8358888 DOI: 10.1111/jcmm.16728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 11/29/2022] Open
Abstract
In recent years, a large number of studies have reported that neuroinflammation aggravates the occurrence of secondary injury after spinal cord injury. Gramine (GM), a natural indole alkaloid, possesses various pharmacological properties; however, the anti-inflammation property remains unclear. In our study, Gramine was investigated in vitro and in vivo to explore the neuroprotection effects. In vitro experiment, our results suggest that Gramine treatment can inhibit release of pro-inflammatory mediators. Moreover, Gramine prevented apoptosis of PC12 cells which was caused by activated HAPI microglia, and the inflammatory secretion ability of microglia was inhibited by Gramine through NF-κB pathway. The in vivo experiment is that 80 mg/kg Gramine was injected orthotopically to rats after spinal cord injury (SCI). Behavioural and histological analyses demonstrated that Gramine treatment may alleviate microglia activation and then boost recovery of motor function after SCI. Overall, our research has demonstrated that Gramine exerts suppressed microglia activation and promotes motor functional recovery after SCI through NF-κB pathway, which may put forward the prospect of clinical treatment of inflammation-related central nervous diseases.
Collapse
Affiliation(s)
- Xiaolang Lu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Fengfeng Lu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Jiachen Yu
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Xinghe Xue
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Hongyi Jiang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Liting Jiang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| | - Yang Yang
- Department of OrthopedicsThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- Zhejiang Provincial Key Laboratory of OrthopedicsWenzhouChina
| |
Collapse
|
47
|
Senent Y, Ajona D, González-Martín A, Pio R, Tavira B. The Complement System in Ovarian Cancer: An Underexplored Old Path. Cancers (Basel) 2021; 13:3806. [PMID: 34359708 PMCID: PMC8345190 DOI: 10.3390/cancers13153806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer is one of the most lethal gynecological cancers. Current therapeutic strategies allow temporary control of the disease, but most patients develop resistance to treatment. Moreover, although successful in a range of solid tumors, immunotherapy has yielded only modest results in ovarian cancer. Emerging evidence underscores the relevance of the components of innate and adaptive immunity in ovarian cancer progression and response to treatment. Particularly, over the last decade, the complement system, a pillar of innate immunity, has emerged as a major regulator of the tumor microenvironment in cancer immunity. Tumor-associated complement activation may support chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and activate cancer-related signaling pathways. Recent insights suggest an important role of complement effectors, such as C1q or anaphylatoxins C3a and C5a, and their receptors C3aR and C5aR1 in ovarian cancer progression. Nevertheless, the implication of these factors in different clinical contexts is still poorly understood. Detailed knowledge of the interplay between ovarian cancer cells and complement is required to develop new immunotherapy combinations and biomarkers. In this context, we discuss the possibility of targeting complement to overcome some of the hurdles encountered in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yaiza Senent
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
| | - Daniel Ajona
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Antonio González-Martín
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Oncology, Clinica Universidad de Navarra, 28027 Madrid, Spain
| | - Ruben Pio
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Beatriz Tavira
- Translational Oncology Group, Program in Solid Tumors, Cima University of Navarra, 31008 Pamplona, Spain; (Y.S.); (A.G.-M.); (R.P.); (B.T.)
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
48
|
Regnier-Golanov AS, Dündar F, Zumbo P, Betel D, Hernandez MS, Peterson LE, Lo EH, Golanov EV, Britz GW. Hippocampal Transcriptome Changes After Subarachnoid Hemorrhage in Mice. Front Neurol 2021; 12:691631. [PMID: 34354664 PMCID: PMC8329593 DOI: 10.3389/fneur.2021.691631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
After subarachnoid hemorrhage (SAH), up to 95% of surviving patients suffer from post-SAH syndrome, which includes cognitive deficits with impaired memory, executive functions, and emotional disturbances. Although these long-term cognitive deficits are thought to result from damage to temporomesial-hippocampal areas, the underlying mechanisms remain unknown. To fill this gap in knowledge, we performed a systematic RNA sequencing screen of the hippocampus in a mouse model of SAH. SAH was induced by perforation of the circle of Willis in mice. Four days later, hippocampal RNA was obtained from SAH and control (sham perforation) mice. Next-generation RNA sequencing was used to determine differentially expressed genes in the whole bilateral hippocampi remote from the SAH bleeding site. Functional analyses and clustering tools were used to define molecular pathways. Differential gene expression analysis detected 642 upregulated and 398 downregulated genes (false discovery rate <0.10) in SAH compared to Control group. Functional analyses using IPA suite, Gene Ontology terms, REACTOME pathways, and MsigDB Hallmark gene set collections revealed suppression of oligodendrocytes/myelin related genes, and overexpression of genes related to complement system along with genes associated with innate and adaptive immunity, and extracellular matrix reorganization. Interferon regulatory factors, TGF-β1, and BMP were identified as major orchestrating elements in the hippocampal tissue response. The MEME-Suite identified binding motifs of Krüppel-like factors, zinc finger transcription factors, and interferon regulatory factors as overrepresented DNA promoter motifs. This study provides the first systematic gene and pathway database of the hippocampal response after SAH. Our findings suggest that damage of the entorhinal cortex by subarachnoid blood may remotely trigger specific hippocampal responses, which include suppression of oligodendrocyte function. Identification of these novel pathways may allow for development of new therapeutic approaches for post-SAH cognitive deficits.
Collapse
Affiliation(s)
- Angelique S. Regnier-Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Friederike Dündar
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
| | - Magda S. Hernandez
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX, United States
| | - Eng H. Lo
- Laboratory of Neuroprotection Research, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Eugene V. Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W. Britz
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
49
|
Yao XQ, Liu ZY, Chen JY, Huang ZC, Liu JH, Sun BH, Zhu QA, Ding RT, Chen JT. Proteomics and bioinformatics reveal insights into neuroinflammation in the acute to subacute phases in rat models of spinal cord contusion injury. FASEB J 2021; 35:e21735. [PMID: 34143440 DOI: 10.1096/fj.202100081rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/18/2021] [Accepted: 06/01/2021] [Indexed: 01/16/2023]
Abstract
Neuroinflammation is recognized as a hallmark of spinal cord injury (SCI). Although neuroinflammation is an important pathogenic factor that leads to secondary injuries after SCI, neuroprotective anti-inflammatory treatments remain ineffective in the management of SCI. Moreover, the molecular signatures involved in the pathophysiological changes that occur during the course of SCI remain ambiguous. The current study investigated the proteins and pathways involved in C5 spinal cord hemi-contusion injury using a rat model by means of 4-D label-free proteomic analysis. Furthermore, two Gene Expression Omnibus (GEO) transcriptomic datasets, Western blot assays, and immunofluorescent staining were used to validate the expression levels and localization of dysregulated proteins. The present study observed that the rat models of SCI were associated with the enrichment of proteins related to the complement and coagulation cascades, cholesterol metabolism, and lysosome pathway throughout the acute and subacute phases of injury. Intriguingly, the current study also observed that 75 genes were significantly altered in both the GEO datasets, including ANXA1, C1QC, CTSZ, GM2A, GPNMB, and PYCARD. Further temporal clustering analysis revealed that the continuously upregulated protein cluster was associated with immune response, lipid regulation, lysosome pathway, and myeloid cells. Additionally, five proteins were further validated by means of Western blot assays and the immunofluorescent staining showed that these proteins coexisted with the F4/80+ reactive microglia and infiltrating macrophages. In conclusion, the proteomic data pertaining to the current study indicate the notable proteins and pathways that may be novel therapeutic targets for the treatment of SCI.
Collapse
Affiliation(s)
- Xin-Qiang Yao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong-Yuan Liu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Ying Chen
- Department of Comprehensive Medical Treatment Ward, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zu-Cheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Hao Liu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Division of Spine Surgery, Department of Orthopaedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Bai-Hui Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qing-An Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruo-Ting Ding
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian-Ting Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Li Z, Zheng M, Yu S, Yao F, Luo Y, Liu Y, Tian D, Cheng L, Jing J. M2 Macrophages Promote PDGFRβ + Pericytes Migration After Spinal Cord Injury in Mice via PDGFB/PDGFRβ Pathway. Front Pharmacol 2021; 12:670813. [PMID: 33935795 PMCID: PMC8082415 DOI: 10.3389/fphar.2021.670813] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/22/2021] [Indexed: 01/06/2023] Open
Abstract
Platelet derived growth factor receptor β positive (PDGFRβ+) pericytes form fibrotic scar, which prevents axonal regeneration after spinal cord injury (SCI). However, the mechanism by which PDGFRβ+ pericytes migrate to the injury core is unclear. Here, we investigated the effect and mechanism of macrophages polarization on PDGFRβ+ pericytes migration after SCI. Macrophages were closely related to the spatiotemporal distribution of PDGFRβ+ pericytes in the injury core at 3, 7, and 14 days postinjury (dpi). Macrophages appeared M2 polarization at 3 and 7 dpi while M1 polarization at 14 dpi. The expression of platelet derived growth factor B (PDGFB) was significantly increased after SCI and after macrophages M2 polarization. The promoting effect of exogenous PDGFB and M2 macrophages conditioned medium on PDGFRβ+ pericytes migration could be blocked by SU16f, a PDGFRβ specific inhibitor. These findings indicate that M2 macrophages can secrete PDGFB acting on PDGFRβ to promote PDGFRβ+ pericytes migration, which can be blocked by a PDGFRβ specific inhibitor SU16f. The PDGFB/PDGFRβ pathway is a promising new target for the treatment of SCI.
Collapse
Affiliation(s)
- Ziyu Li
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Meige Zheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Shuisheng Yu
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Fei Yao
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yang Luo
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yanchang Liu
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Li Cheng
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|