1
|
Zhou HY, Wang X, Li Y, Wang D, Zhou XZ, Xiao N, Li GX, Li G. Dynamic development of microglia and macrophages after spinal cord injury. Neural Regen Res 2025; 20:3606-3619. [PMID: 39101644 PMCID: PMC11974661 DOI: 10.4103/nrr.nrr-d-24-00063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/09/2024] [Accepted: 05/28/2024] [Indexed: 08/06/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00029/figure1/v/2025-01-31T122243Z/r/image-tiff Secondary injury following spinal cord injury is primarily characterized by a complex inflammatory response, with resident microglia and infiltrating macrophages playing pivotal roles. While previous studies have grouped these two cell types together based on similarities in structure and function, an increasing number of studies have demonstrated that microglia and macrophages exhibit differences in structure and function and have different effects on disease processes. In this study, we used single-cell RNA sequencing and spatial transcriptomics to identify the distinct evolutionary paths of microglia and macrophages following spinal cord injury. Our results showed that microglia were activated to a pro-inflammatory phenotype immediately after spinal cord injury, gradually transforming to an anti-inflammatory steady state phenotype as the disease progressed. Regarding macrophages, our findings highlighted abundant communication with other cells, including fibroblasts and neurons. Both pro-inflammatory and neuroprotective effects of macrophages were also identified; the pro-inflammatory effect may be related to integrin β2 ( Itgb2 ) and the neuroprotective effect may be related to the oncostatin M pathway. These findings were validated by in vivo experiments. This research underscores differences in the cellular dynamics of microglia and macrophages following spinal cord injury, and may offer new perspectives on inflammatory mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Hu-Yao Zhou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xia Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Yi Li
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Duan Wang
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Xuan-Zi Zhou
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Nong Xiao
- Department of Rehabilitation, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - Guo-Xing Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Li P, Liu J, Wang Y, Li M, Gong X, Peng Z, Liu Y, Zhang Y, Luan Z, Liu D, Wang Y. Spatiotemporal Targeted Delivery of Biomimetic Bacterial Outer Membrane Nanoparticles for Enhanced Spinal Cord Injury Repair. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2502795. [PMID: 40391641 DOI: 10.1002/adma.202502795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/30/2025] [Indexed: 05/22/2025]
Abstract
Spinal cord injury (SCI) is a complex and dynamic pathological condition characterized by disrupted lipid metabolism and neuroinflammatory responses, posing significant therapeutic challenges. To address these, biomimetic bacterial outer membrane nanoparticles (BM-NPs) are designed by integrating the precise targeting capability of detoxified outer membrane vesicles (dOMVs) with the efficient drug-loading properties of liposomes. BM-NPs exhibit superior targeting efficiency toward peripheral neutrophils and macrophages, enabling spatiotemporal drug delivery via immune cells. An innovative "Tortoise and Hare" dynamic adaptive delivery strategy is introduced, where neutrophils facilitate rapid drug transport during the acute phase of SCI, while macrophages ensure sustained delivery during the subacute phase. This strategy aligns with the dynamic pathological progression of SCI, offering precision targeting tailored to different stages of injury. BM-NPs demonstrate multifaceted therapeutic effects, including the suppression of foam cell formation through coordinated enhancement of lipid droplet autophagy and cholesterol efflux. Furthermore, they modulate the inflammatory microenvironment, preserve myelin integrity, and significantly promote neural functional recovery post-SCI. By overcoming the limitations of conventional delivery systems in targeting and timeliness, BM-NPs offer an innovative, highly efficient, and clinically translatable platform for SCI treatment and other acute inflammatory disorders of the central nervous system.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Jingsong Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Yangyang Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Mi Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Xuqiang Gong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Zhibin Peng
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150000, P. R. China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin, Heilongjiang, 150000, P. R. China
| | - Yishu Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Yubo Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Zhiwei Luan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| | - Daqian Liu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Yansong Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150000, P. R. China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150000, P. R. China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, 150000, P. R. China
| |
Collapse
|
3
|
Saraswat Ohri S, Myers SA, Rood B, Brown BL, Chilton PM, Slomnicki L, Liu Y, Wei GZ, Andres KR, Mohan D, Howard RM, Whittemore SR, Hetman M. Reduced White Matter Damage and Lower Neuroinflammatory Potential of Microglia and Macrophages in Hri/Eif2ak1 -/- Mice After Contusive Spinal Cord Injury. Glia 2025; 73:1004-1021. [PMID: 39760211 DOI: 10.1002/glia.24669] [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: 05/22/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
Cellular stressors inhibit general protein synthesis while upregulating stress response transcripts and/or proteins. Phosphorylation of the translation factor eIF2α by one of the several stress-activated kinases is a trigger for such signaling, known as the integrated stress response (ISR). The ISR regulates cell survival and function under stress. Here, germline knockout mice were used to determine contributions by three major ISR kinases, HRI/EIF2AK1, GCN2/EIF2AK4, and PKR//EIF2AK2, to pathogenesis of moderate contusive spinal cord injury (SCI) at the thoracic T9 level. One-day post-injury (dpi), reduced levels of peIF2α were found in Hri -/- and Gcn2 -/-, but not in Pkr -/- mice. In addition, Hri -/- mice showed attenuated expression of the downstream ISR transcripts, Atf4 or Chop. Such differential effects of SCI-activated ISR correlated with a strong or moderate enhancement of locomotor recovery in Hri -/- or Gcn2 -/- mice, respectively. Hri -/- mice also showed reduced white matter loss, increased content of oligodendrocytes (OL) and attenuated neuroinflammation, including decreased lipid accumulation in microglia/macrophages. Cultured neonatal Hri -/- OLs showed lower ISR cytotoxicity. Moreover, cell autonomous reduction in neuroinflammatory potential was observed in microglia and bone marrow-derived macrophages derived from Hri -/- mice. These data identify HRI as a major positive regulator of SCI-associated secondary injury. In addition, targeting HRI may enable multimodal neuroprotection to enhance functional recovery after SCI.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Scott A Myers
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Benjamin Rood
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Brandon L Brown
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Paula M Chilton
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Norton Neuroscience Institute, Louisville, Kentucky, USA
| | - Lukasz Slomnicki
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Yu Liu
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - George Z Wei
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Divya Mohan
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Biochemistry & Molecular Genetics, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Pharmacology & Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Department of Interdisciplinary Program in Translational Neuroscience, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
4
|
Yan R, Yuan Y, Shi C, Li Y, Li Y, Wang W, Yang L. Kanglexin attenuates spinal cord injury by modulating pyroptosis and polarization via the PKA/NF-κB signaling pathway. Int Immunopharmacol 2025; 153:114401. [PMID: 40101425 DOI: 10.1016/j.intimp.2025.114401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Neuroinflammation is essential for intricate pathophysiologic mechanisms after spinal cord injury (SCI). Increasing evidence suggests that anthraquinones possess anti-inflammatory properties in central nervous system (CNS) disorders. However, the effects of Kanglexin (Klx), a novel synthetic anthraquinone compound, on SCI remain unknown. METHODS C57BL/6 mice were utilized to establish a contused SCI model to explore the in vivo neuroprotective and inflammatory modulatory effects of Klx. An inflammation model was also created in vitro using BV2 cells. Neuroprotective effects were assessed by evaluating motor function and neuropathologic alterations. Inflammation modulation was analyzed through markers of polarization and pyroptosis, with further mechanistic insights obtained via transcriptome sequencing. RESULTS Klx facilitated the recovery of hindlimb locomotor function and improved neuronal survival after SCI. Both in vitro and in vivo assays revealed that Klx inhibited NLRP3 inflammasome-induced pyroptosis. In addition, Klx promoted the polarization of microglia from the proinflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Mechanistically, Klx enhanced PKA phosphorylation and suppressed NF-κB and IκBα phosphorylation, thereby reducing NF-κB nuclear translocation. CONCLUSION Klx demonstrated neuroprotective and inflammation-modulating effects on SCI, suggesting that it might offer a promising therapeutic alternative for SCI.
Collapse
Affiliation(s)
- Rongbao Yan
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ye Yuan
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ce Shi
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Yang Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China; NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin, China; Joint Key Laboratory of Endemic Diseases(Harbin Medical University, Guizhou Medical University, Xi'an Jiaotong University), Harbin Medical University, Harbin Medical University, Harbin, China; Heilongjiang Provincial Key Laboratory of Trace Elements and Human Health, Harbin Medical University, Harbin, China; Key Laboratory of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Harbin Medical University, Harbin, China.
| | - Yang Li
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery of Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Liu J, Huang Z, Chen K, Li R, Huang Z, Lin J, Jiang H, Liu J, Zhu Q. Histopathological Insights into Demyelination and Remyelination After Spinal Cord Injury in Non-human Primates. Neurosci Bull 2025:10.1007/s12264-025-01388-2. [PMID: 40186822 DOI: 10.1007/s12264-025-01388-2] [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: 08/28/2024] [Accepted: 01/08/2025] [Indexed: 04/07/2025] Open
Abstract
Demyelination and remyelination play key roles in spinal cord injury (SCI), affecting the recovery of motor and sensory functions. Research in rodent models is extensive, but the study of these processes in non-human primates is limited. Therefore, our goal was to thoroughly study the histological features of demyelination and remyelination after contusion injury of the cervical spinal cord in Macaca fascicularis. In a previous study, we created an SCI model in M. fascicularis by controlling the contusion displacement. We used Eriochrome Cyanine staining, immunohistochemical analysis, and toluidine blue staining to evaluate demyelination and remyelination. The results showed demyelination ipsilateral to the injury epicenter both rostrally and caudally, the former mainly impacting sensory pathways, while the latter primarily affected motor pathways. Toluidine blue staining showed myelin loss and axonal distension at the injury site. Schwann cell-derived myelin sheaths were only found at the center, while thinner myelin sheaths from oligodendrocytes were seen at the center and surrounding areas. Our study showed that long-lasting demyelination occurs in the spinal cord of M. fascicularis after SCI, with oligodendrocytes and Schwann cells playing a significant role in myelin sheath formation at the injury site.
Collapse
Affiliation(s)
- Junhao Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Division of Spinal Surgery, Department of Orthopaedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510000, China
| | - Zucheng Huang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kinon Chen
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Rong Li
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiping Huang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Junyu Lin
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hui Jiang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jie Liu
- Institute of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China.
| | - Qingan Zhu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Wu W, Meng T, Wang Y, Chen J, Liang C. Prdx5 regulates macrophage polarization by modulating the TLR4/NF-κB pathway to promote apoptosis in chronic prostatitis. Int Immunopharmacol 2025; 151:114332. [PMID: 40015209 DOI: 10.1016/j.intimp.2025.114332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a prevalent urological disorder characterized by urinary symptoms, pelvic pain, and sexual dysfunction. Despite its high prevalence, the pathogenesis of CP/CPPS remains poorly understood. Our study revealed that peroxiredoxin 5 (Prdx5) was upregulated in M1 macrophages and in mice with experimental autoimmune prostatitis (EAP), with its expression in macrophages being regulated in a reactive oxygen species (ROS)-dependent manner. Using western blotting, RT-qPCR, immunohistochemical staining, hematoxylin and eosin staining, immunofluorescence staining, flow cytometry, and cell co-culturing, it was demonstrated that the silencing of Prdx5 suppressed the polarization of macrophages towards the M1 phenotype. This inhibition reduced apoptosis in prostate epithelial cells and mitigated the progression of EAP. Furthermore, Prdx5 mediated its effects in macrophages and EAP via the Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) pathway. Our findings suggest that Prdx5 promoted the occurrence and development of CP/CPPS due to its promotion of M1 polarization and apoptosis of prostate epithelial cells in an ROS-dependent manner via the TLR4/NF-κB axis, indicating its potential as a therapeutic target to treat CP/CPPS.
Collapse
Affiliation(s)
- Weikang Wu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Tong Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Yufan Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China.
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology, and Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei 230022, Anhui, China.
| |
Collapse
|
7
|
Zhang C, Zhao S, Huang Z, Xue A, Liu H, Dai S, Zheng Z, Li Y, Guo X, Gu J, Zhang F, Wang F, Wang Y, Zhou X, Zhang S, Zhang H, Shen J, Chen J, Yin G. Macropinocytosis enhances foamy macrophage formation and cholesterol crystallization to activate NLRP3 inflammasome after spinal cord injury. Redox Biol 2025; 79:103469. [PMID: 39700693 PMCID: PMC11723182 DOI: 10.1016/j.redox.2024.103469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/30/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
After spinal cord injury (SCI), phagocytes endocytose myelin debris to form foam cells, exacerbating the inflammatory response. It has been previously shown that macrophages become foam cells through the phagocytosis of myelin debris via receptor-dependent mechanisms after SCI. Blocking receptor-mediated endocytosis did not completely prevent foam cell formation, so we investigated receptor-independent endocytosis. Here, we revealed that foam cells formed after myelin debris internalization were predominantly macrophages rather than microglia. Receptor-independent macropinocytosis has an important position in foamy macrophage formation through engagement of myelin debris endocytosis after SCI. Mechanistic studies showed that cholesterol crystallization following macropinocytosis-mediated foamy macrophage formation promoted the reactive oxygen species (ROS) production and the NOD-like receptor protein 3 (NLRP3) inflammasome activation, increasing the secretion of interleukin-1β (IL-1β). Inhibition of macropinocytosis might reverse this effect, resulting in enhanced axonal regeneration and reduced neural apoptosis, thereby improving outcomes after SCI. Overall, our study revealed a previously unrecognized role for macropinocytosis in foamy macrophages formation after SCI, and confer a promising therapeutic strategy for SCI through focus on macropinocytosis.
Collapse
Affiliation(s)
- Chenxi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China; Department of Orthopedics, Suzhou Municipal Hospital, Nanjing Medical University, Jiangsu, Suzhou, 215000, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Ao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Siming Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Ziyang Zheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Yin Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Xiaodong Guo
- Department of Orthopedics, Wuhan Union Hospital of China, Huazhong University of Science and Technology, Hubei, Wuhan, 430022, China
| | - Jun Gu
- Department of Orthopedics, Xishan People's Hospital of Wuxi City, Southeast University, Jiangsu, Wuxi, 21405, China
| | - Feng Zhang
- Department of Orthopedics, Xuyi People's Hospital, Yangzhou University, Jiangsu, Xuyi, 211700, China
| | - Fubing Wang
- Department of Orthopedics, Xuyi People's Hospital, Yangzhou University, Jiangsu, Xuyi, 211700, China
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, Nanjing Medical University, Jiangsu, Yangzhou, 225001, China
| | - Xiaohua Zhou
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China
| | - Shujun Zhang
- Department of Orthopedics, Wuxi Ninth Hospital, Suzhou University, Jiangsu, Wuxi, 21405, China
| | - Hanwen Zhang
- Department of Pathophysiology, Nanjing Medical University, Jiangsu, Nanjing, 211166, China.
| | - Jun Shen
- Department of Orthopedics, Suzhou Municipal Hospital, Nanjing Medical University, Jiangsu, Suzhou, 215000, China.
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, Nanjing, 210029, China.
| |
Collapse
|
8
|
Ma Z, Meng C, Wang X, Zhao Y, Wang J, Chen Y, Li Y, Jiang Y, Ouyang F, Li J, Zheng M, Cheng L, Jing J. Trehalose enhances macrophage autophagy to promote myelin debris clearance after spinal cord injury. Cell Biosci 2025; 15:11. [PMID: 39881390 PMCID: PMC11781065 DOI: 10.1186/s13578-025-01357-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Myelin-laden foamy macrophages accumulate extensively in the lesion epicenter, exhibiting characteristics of autophagolysosomal dysfunction, which leads to prolonged inflammatory responses after spinal cord injury (SCI). Trehalose, known for its neuroprotective properties as an autophagy inducer, has yet to be fully explored for its potential to mitigate foamy macrophage formation and exert therapeutic effects in the context of SCI. RESULTS We observed that trehalose significantly enhances macrophage phagocytosis and clearance of myelin in a dose-dependent manner in vitro. In vivo, trehalose administration markedly reduced myelin debris accumulation, inhibited foamy macrophage formation, suppressed inflammatory responses, decreased fibrotic scarring, and promoted axonal growth and motor function recovery after SCI. These beneficial effects of trehalose may be related to the overexpression of transcription factor EB (TFEB), a key regulator of the autophagy-lysosomal system, which can rescue autophagic dysfunction in foamy macrophages and inhibit inflammatory responses. Additionally, the effects of trehalose on macrophages were abolished by chloroquine, an autophagy inhibitor, suggesting trehalose's potential as a therapeutic candidate for enhancing myelin debris clearance post-SCI. CONCLUSIONS Our findings underscore the pivotal role of trehalose in modulating myelin debris clearance within macrophages, providing new perspectives for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Zhida Ma
- 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
| | - Congpeng Meng
- 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
| | - Xiang 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
| | - 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
| | - Yihao 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
| | - Yiteng 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
| | - Yan Jiang
- Department of Rehabilitation 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
| | - 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.
| | - 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.
| | - 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.
| |
Collapse
|
9
|
Yang X, Tang C, Zhu D, Xia X, Du Q, Huang L, Liu J, Liu Y. Nonylphenol exposure increases the risk of Hirschsprung's disease by inducing macrophage M1 polarization. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117756. [PMID: 39837008 DOI: 10.1016/j.ecoenv.2025.117756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Nonylphenol (NP), a ubiquitous environmental contaminant used as a surfactant in industrial production and classified as an endocrine disruptor, could interfere hormone secretion and exhibit neurotoxicity in organisms. Hirschsprung's disease (HSCR), one of the most frequently observed congenital malformations of the digestive system, arises mainly due to the failure of enteric neural crest cells to migrate to the distal colon during embryonic development. However, the effects of NP exposure on HSCR are largely unknown. Herein, we identified the content of NP and expression of lncRNA LINC00294/Inhibin Subunit Beta E (INHBE) axis in clinical samples and evaluated the crucial role of lncRNA LINC00294/INHBE axis in the neurogenic potential of neurons and the neurotoxicity effects of NP in the SH-SY5Y cells and female specific pathogen-free (SPF) rat model. Our results showed that NP concentration and LINC00294 were significantly associated with HSCR occurrence and macrophage polarization in human HSCR specimens. Moreover, NP promoted macrophage M1 polarization. The proliferation and migration were weakened, and apoptosis was heightened by the conditioned medium of NP-treated macrophages in SH-SY5Y cells. Contrarily, LINC00294 overexpression and INHBE knockdown reversed the neurotoxicity effect of NP on SH-SY5Y cells. Furthermore, the neurotoxicity effect of NP was abolished by clodronate liposomes in the rat model. In conclusion, NP could induce macrophage M1 polarization via the LINC00294/INHBE axis and increase the risk of Hirschsprung's disease. Our findings would provide a foundation for the toxicity study and risk assessments of NP.
Collapse
Affiliation(s)
- Xuefeng Yang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China; Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chengyan Tang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Daiwei Zhu
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Xingrong Xia
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Qing Du
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Lu Huang
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China
| | - Jianguo Liu
- Key Laboratory of Oral Disease Research, School of Stomatology, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Yuanmei Liu
- Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Pediatric Surgery, Guizhou Children Hospital, Zunyi, Guizhou 563000, China.
| |
Collapse
|
10
|
Li J, Zheng M, Ouyang F, Ye J, Huang J, Zhao Y, Wang J, Shan F, Li Z, Yu S, Yao F, Tian D, Cheng L, Jing J. Interleukin-3 Modulates Macrophage Phagocytic Activity and Promotes Spinal Cord Injury Repair. CNS Neurosci Ther 2024; 30:e70181. [PMID: 39697159 DOI: 10.1111/cns.70181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/28/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Effective clearance of lipid-rich debris by macrophages is critical for neural repair and regeneration after spinal cord injury (SCI). Interleukin-3 (IL-3) has been implicated in programming microglia to cluster and clear pathological aggregates in neurodegenerative disease. Yet, the influence of IL-3 on lipid debris clearance post-SCI is not well characterized. METHODS We established a mouse model of spinal cord compression injury to investigate the role of IL-3. Blockage of IL-3 was achieved through intrathecal delivery of an IL-3-neutralizing antibody, while IL-3 activation was augmented via in situ injection of recombinant IL-3 into the lesion site immediately post-SCI. Immunofluorescence staining was performed to determine IL-3 and IL-3Rα sources and distribution, lipid droplet accumulation, neuron preservation, and axon regeneration after SCI. The Basso Mouse Scale (BMS) and footprint analysis were employed to evaluate locomotor function recovery. RESULTS We found that IL-3 expression was significantly upregulated post-SCI, peaking at 14 days post-injury (dpi) and persisting until 28 dpi. Notably, IL-3 was primarily secreted by astrocytes surrounding the lesion epicenter. Correspondingly, IL-3Rα was predominantly observed in macrophages within the injury core, also elevating at 14 dpi. Neutralization of IL-3 led to increased lipid droplet accumulation, along with markedly widespread of macrophages and decreased neuronal survival, resulting in severe motor deficits compared to controls. Conversely, in situ injection of IL-3 reduced lipid droplet accumulation in macrophages, preserved neurons, promoted axon regeneration, and ultimately contributed to the recovery of motor function after SCI. CONCLUSION Our findings shed light on the role of IL-3 in modulating macrophage phagocytic activity and suggest that the IL-3/IL-3Rα pathway may be a potential therapeutic target for enhancing neural repair and functional recovery after SCI.
Collapse
Affiliation(s)
- Jianjian Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Meige Zheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangru Ouyang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianan Ye
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinxin Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanzhe Zhao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingwen Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangli Shan
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyu Li
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuishen Yu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fei Yao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dasheng Tian
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Cheng
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juehua Jing
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
11
|
Ding R, Wu L, Wei S, Lu H, Qin X, Liu X, Wang Y, Liu W, Li H, Luo B, Xie T, Chen Z. Multi-targeted olink proteomics analyses of cerebrospinal fluid from patients with aneurysmal subarachnoid hemorrhage. Proteome Sci 2024; 22:11. [PMID: 39604965 PMCID: PMC11600900 DOI: 10.1186/s12953-024-00236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The complexity of delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (aSAH) may require the simultaneous analysis of variant types of protein biomarkers to describe it more accurately. In this study, we analyzed for the first time the alterations of cerebrospinal fluid (CSF) proteins in patients with aSAH by multi-targeted Olink proteomics, aiming to reveal the pathophysiology of DCI and provide insights into the diagnosis and treatment of aSAH. METHODS Six aSAH patients and six control patients were selected, and CSF samples were analyzed by Olink Proteomics (including 96-neurology panel and 96-inflammation panel) based on Proximity Extension Assay (PEA). Differentially expressed proteins (DEPs) were acquired and bioinformatics analysis was performed. RESULTS PCA analysis revealed better intra- and inter-group reproducibility of CSF samples in the control and aSAH groups. 23 neurology-related and 31 inflammation-relevant differential proteins were identified. In the neurology panel, compared to controls, the up-regulated proteins in the CSF of SAH patients predominantly included macrophage scavenger receptor 1 (MSR1), siglec-1, siglec-9, cathepsin C (CTSC), cathepsin S (CTSS), etc. Meanwhile, in the inflammation group, the incremental proteins mainly contained interleukin-6 (IL-6), MCP-1, CXCL10, CXCL-9, TRAIL, etc. Cluster analysis exhibited significant differences in differential proteins between the two groups. GO function enrichment analysis hinted that the differential proteins pertinent to neurology in the CSF of SAH patients were mainly involved in the regulation of defense response, vesicle-mediated transport and regulation of immune response; while the differential proteins related to inflammation were largely connected with the cellular response to chemokine, response to chemokine and chemokine-mediated signaling pathway. Additionally, in the neurology panel, KEGG enrichment analysis indicated that the differential proteins were significantly enriched in the phagosome, apoptosis and microRNAs in cancer pathway. And in the inflammation panel, the differential proteins were mainly enriched in the chemokine signaling pathway, viral protein interaction with cytokine and cytokine receptor and toll-like receptor signaling pathway. CONCLUSIONS These identified differential proteins reveal unique pathophysiological characteristics secondary to aSAH. Further characterization of these proteins and aberrant pathways in future research could enable their application as potential therapeutic targets and biomarkers for DCI after aSAH.
Collapse
Affiliation(s)
- Rui Ding
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Liquan Wu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Shanshan Wei
- Department of Oncology, Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Haoran Lu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Xiaohong Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Xizhi Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China
| | - Yanhua Wang
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Wen Liu
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Huibing Li
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Baochang Luo
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China
| | - Teng Xie
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China.
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Street, Wuhan, 430060, China.
- Department of Neurosurgery, Hanchuan Renmin Hospital, Hanchuan, Hubei, 431600, China.
| |
Collapse
|
12
|
Zhou Y, Xu T, Zhou Y, Han W, Wu Z, Yang C, Chen X. A review focuses on a neglected and controversial component of SCI: myelin debris. Front Immunol 2024; 15:1436031. [PMID: 39650659 PMCID: PMC11621000 DOI: 10.3389/fimmu.2024.1436031] [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: 05/21/2024] [Accepted: 10/22/2024] [Indexed: 12/11/2024] Open
Abstract
Myelin sheath, as the multilayer dense structure enclosing axons in humans and other higher organisms, may rupture due to various injury factors after spinal cord injury, thus producing myelin debris. The myelin debris contains a variety of myelin-associated inhibitors (MAIs) and lipid, all inhibiting the repair after spinal cord injury. Through summary and analysis, the present authors found that the inhibition of myelin debris can be mainly divided into two categories: firstly, the direct inhibition mediated by MAIs; secondly, the indirect inhibition mediated by lipid such as cholesterol. It is worth noting that phagocytes are required in the latter indirect inhibition, such as professional phagocytes (macrophages et al.) and non-professional phagocytes (astrocytes et al.). Moreover, complement and the immune system also participate in the phagocytosis of myelin debris, working together with phagocytes to aggravate spinal cord injury. In conclusion, this paper focuses on the direct and indirect effects of myelin debris on spinal cord injury, aiming to provide new inspiration and reflection for the basic research of spinal cord injury and the conception of related treatment.
Collapse
Affiliation(s)
- Yuchen Zhou
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Tao Xu
- Medical School of Nantong University, Nantong, China
- Department of Orthopedics, Yancheng Dafeng People's Hospital, Yancheng, China
| | - Yiyan Zhou
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Wei Han
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Zhengchao Wu
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Changwei Yang
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Xiaoqing Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
13
|
Gao R, Song SJ, Tian MY, Wang LB, Zhang Y, Li X. Myelin debris phagocytosis in demyelinating disease. Glia 2024; 72:1934-1954. [PMID: 39073200 DOI: 10.1002/glia.24602] [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: 12/26/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Demyelinating diseases are often caused by a variety of triggers, including immune responses, viral infections, malnutrition, hypoxia, or genetic factors, all of which result in the loss of myelin in the nervous system. The accumulation of myelin debris at the lesion site leads to neuroinflammation and inhibits remyelination; therefore, it is crucial to promptly remove the myelin debris. Initially, Fc and complement receptors on cellular surfaces were the primary clearance receptors responsible for removing myelin debris. However, subsequent studies have unveiled the involvement of additional receptors, including Mac-2, TAM receptors, and the low-density lipoprotein receptor-related protein 1, in facilitating the removal process. In addition to microglia and macrophages, which serve as the primary effector cells in the disease phase, a variety of other cell types such as astrocytes, Schwann cells, and vascular endothelial cells have been demonstrated to engage in the phagocytosis of myelin debris. Furthermore, we have concluded that oligodendrocyte precursor cells, as myelination precursor cells, also exhibit this phagocytic capability. Moreover, our research group has innovatively identified the low-density lipoprotein receptor as a potential phagocytic receptor for myelin debris. In this article, we discuss the functional processes of various phagocytes in demyelinating diseases. We also highlight the alterations in signaling pathways triggered by phagocytosis, and provide a comprehensive overview of the various phagocytic receptors involved. Such insights are invaluable for pinpointing potential therapeutic strategies for the treatment of demyelinating diseases by targeting phagocytosis.
Collapse
Affiliation(s)
- Rui Gao
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Sheng-Jiao Song
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Meng-Yuan Tian
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Li-Bin Wang
- Neurosurgery Department, Huazhong University of Science and Technology Union Shenzhen Hospital/Shenzhen Nanshan Hospital, Shenzhen, Guangdong, China
| | - Yuan Zhang
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xing Li
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
14
|
Liu H, Yi J, Zhang C, Li Y, Wang Q, Wang S, Dai S, Zheng Z, Jiang T, Gao P, Xue A, Huang Z, Kong F, Wang Y, He B, Guo X, Li Q, Chen J, Yin G, Zhao S. Macrophage GIT1 promotes oligodendrocyte precursor cell differentiation and remyelination after spinal cord injury. Glia 2024; 72:1674-1692. [PMID: 38899731 DOI: 10.1002/glia.24577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
Spinal cord injury (SCI) can result in severe motor and sensory deficits, for which currently no effective cure exists. The pathological process underlying this injury is extremely complex and involves many cell types in the central nervous system. In this study, we have uncovered a novel function for macrophage G protein-coupled receptor kinase-interactor 1 (GIT1) in promoting remyelination and functional repair after SCI. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we identified that GIT1 deficiency in macrophages led to an increased generation of tumor necrosis factor-alpha (TNFα), reduced proportion of mature oligodendrocytes (mOLs), impaired remyelination, and compromised functional recovery in vivo. These effects in GIT1 CKO mice were reversed with the administration of soluble TNF inhibitor. Moreover, bone marrow transplantation from GIT1 CWT mice reversed adverse outcomes in GIT1 CKO mice, further indicating the role of macrophage GIT1 in modulating spinal cord injury repair. Our in vitro experiments showed that macrophage GIT1 plays a critical role in secreting TNFα and influences the differentiation of oligodendrocyte precursor cells (OPCs) after stimulation with myelin debris. Collectively, our data uncovered a new role of macrophage GIT1 in regulating the transformation of OPCs into mOLs, essential for functional remyelination after SCI, suggesting that macrophage GIT1 could be a promising treatment target of SCI.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Yi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Orthopedics, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Chenxi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yin Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shenyu Wang
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Siming Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyang Zheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fanqi Kong
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Baorong He
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong Uinversity, School of Medicine, Xi'an, China
| | - Xiaodong Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
15
|
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
|
16
|
Li F, Sun X, Sun K, Kong F, Jiang X, Kong Q. Lupenone improves motor dysfunction in spinal cord injury mice through inhibiting the inflammasome activation and pyroptosis in microglia via the nuclear factor kappa B pathway. Neural Regen Res 2024; 19:1802-1811. [PMID: 38103247 PMCID: PMC10960275 DOI: 10.4103/1673-5374.389302] [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: 09/22/2022] [Revised: 07/27/2023] [Accepted: 09/13/2023] [Indexed: 12/18/2023] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202408000-00034/figure1/v/2023-12-16T180322Z/r/image-tiff Spinal cord injury-induced motor dysfunction is associated with neuroinflammation. Studies have shown that the triterpenoid lupenone, a natural product found in various plants, has a remarkable anti-inflammatory effect in the context of chronic inflammation. However, the effects of lupenone on acute inflammation induced by spinal cord injury remain unknown. In this study, we established an impact-induced mouse model of spinal cord injury, and then treated the injured mice with lupenone (8 mg/kg, twice a day) by intraperitoneal injection. We also treated BV2 cells with lipopolysaccharide and adenosine 5'-triphosphate to simulate the inflammatory response after spinal cord injury. Our results showed that lupenone reduced IκBα activation and p65 nuclear translocation, inhibited NLRP3 inflammasome function by modulating nuclear factor kappa B, and enhanced the conversion of proinflammatory M1 microglial cells into anti-inflammatory M2 microglial cells. Furthermore, lupenone decreased NLRP3 inflammasome activation, NLRP3-induced microglial cell polarization, and microglia pyroptosis by inhibiting the nuclear factor kappa B pathway. These findings suggest that lupenone protects against spinal cord injury by inhibiting inflammasomes.
Collapse
Affiliation(s)
- Fudong Li
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Jiang
- Department of Anesthesiology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qingjie Kong
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Qi Y, Zhang Y, Guan S, Liu L, Wang H, Chen Y, Zhou Q, Xu F, Zhang Y. Common ground on immune infiltration landscape and diagnostic biomarkers in diabetes-complicated atherosclerosis: an integrated bioinformatics analysis. Front Endocrinol (Lausanne) 2024; 15:1381229. [PMID: 39145311 PMCID: PMC11323117 DOI: 10.3389/fendo.2024.1381229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/25/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) is a major cause of atherosclerosis (AS). However, definitive evidence regarding the common molecular mechanisms underlying these two diseases are lacking. This study aimed to investigate the mechanisms underlying the association between T2DM and AS. Methods The gene expression profiles of T2DM (GSE159984) and AS (GSE100927) were obtained from the Gene Expression Omnibus, after which overlapping differentially expressed gene identification, bioinformatics enrichment analyses, protein-protein interaction network construction, and core genes identification were performed. We confirmed the discriminatory capacity of core genes using receiver operating curve analysis. We further identified transcription factors using TRRUST database to build a transcription factor-mRNA regulatory network. Finally, the immune infiltration and the correlation between core genes and differential infiltrating immune cells were analyzed. Results A total of 27 overlapping differentially expressed genes were identified under the two-stress conditions. Functional analyses revealed that immune responses and transcriptional regulation may be involved in the potential pathogenesis. After protein-protein interaction network deconstruction, external datasets, and qRT-PCR experimental validation, four core genes (IL1B, C1QA, CCR5, and MSR1) were identified. ROC analysis further showed the reliable value of these core genes. Four common differential infiltrating immune cells (B cells, CD4+ T cells, regulatory T cells, and M2 macrophages) between T2DM and AS datasets were selected based on immune cell infiltration. A significant correlation between core genes and common differential immune cells. Additionally, five transcription factors (RELA, NFκB1, JUN, YY1, and SPI1) regulating the transcription of core genes were mined using upstream gene regulator analysis. Discussion In this study, common target genes and co-immune infiltration landscapes were identified between T2DM and AS. The relationship among five transcription factors, four core genes, and four immune cells profiles may be crucial to understanding T2DM complicated with AS pathogenesis and therapeutic direction.
Collapse
Affiliation(s)
- Yifei Qi
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Zhang
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuang Guan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongqin Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Chen
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Wang X, Cheng Z, Tai W, Shi M, Ayazi M, Liu Y, Sun L, Yu C, Fan Z, Guo B, He X, Sun D, Young W, Ren Y. Targeting foamy macrophages by manipulating ABCA1 expression to facilitate lesion healing in the injured spinal cord. Brain Behav Immun 2024; 119:431-453. [PMID: 38636566 DOI: 10.1016/j.bbi.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.
Collapse
Affiliation(s)
- Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| | - Zhijian Cheng
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Wenjiao Tai
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Mingjun Shi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Yang Liu
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Caiyong Yu
- Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an 710032, China
| | - Bin Guo
- Department of Pathology, Guizhou Medical University, Guiyang 550025, China
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Yi Ren
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
19
|
Kong F, Yu H, Gao L, Xing E, Yu Y, Sun X, Wang W, Zhao D, Li X. Multifunctional Hierarchical Nanoplatform with Anisotropic Bimodal Mesopores for Effective Neural Circuit Reconstruction after Spinal Cord Injury. ACS NANO 2024; 18:13333-13345. [PMID: 38717602 DOI: 10.1021/acsnano.4c03252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
A persistent inflammatory response, intrinsic limitations in axonal regenerative capacity, and widespread presence of extrinsic axonal inhibitors impede the restoration of motor function after a spinal cord injury (SCI). A versatile treatment platform is urgently needed to address diverse clinical manifestations of SCI. Herein, we present a multifunctional nanoplatform with anisotropic bimodal mesopores for effective neural circuit reconstruction after SCI. The hierarchical nanoplatform features of a Janus structure consist of dual compartments of hydrophilic mesoporous silica (mSiO2) and hydrophobic periodic mesoporous organosilica (PMO), each possessing distinct pore sizes of 12 and 3 nm, respectively. Unlike traditional hierarchical mesoporous nanomaterials with dual-mesopores interlaced with each other, the two sets of mesopores in this Janus nanoplatform are spatially independent and possess completely distinct chemical properties. The Janus mesopores facilitate controllable codelivery of dual drugs with distinct properties: the hydrophilic macromolecular enoxaparin (ENO) and the hydrophobic small molecular paclitaxel (PTX). Anchoring with CeO2, the resulting mSiO2&PMO-CeO2-PTX&ENO nanoformulation not only effectively alleviates ROS-induced neuronal apoptosis but also enhances microtubule stability to promote intrinsic axonal regeneration and facilitates axonal extension by diminishing the inhibitory effect of extracellular chondroitin sulfate proteoglycans. We believe that this functional dual-mesoporous nanoplatform holds significant potential for combination therapy in treating severe multifaceted diseases.
Collapse
Affiliation(s)
- Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Hongyue Yu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Lifei Gao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Enyun Xing
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Yan Yu
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Spine Center, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Wenxing Wang
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Dongyuan Zhao
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| | - Xiaomin Li
- Department of Chemistry, Shanghai Stomatological Hospital & School of Stomatology, State Key Laboratory of Molecular Engineering of Polymers, iChem, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
20
|
Dong Y, Kang Z, Zhang Z, Zhang Y, Zhou H, Liu Y, Shuai X, Li J, Yin L, Wang X, Ma Y, Fan H, Jiang F, Lin Z, Ding C, Yun Jin K, Sarapultsev A, Li F, Zhang G, Xie T, Yin C, Cheng X, Luo S, Liu Y, Hu D. Single-cell profile reveals the landscape of cardiac immunity and identifies a cardio-protective Ym-1 hi neutrophil in myocardial ischemia-reperfusion injury. Sci Bull (Beijing) 2024; 69:949-967. [PMID: 38395651 DOI: 10.1016/j.scib.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/11/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a major hindrance to the success of cardiac reperfusion therapy. Although increased neutrophil infiltration is a hallmark of MIRI, the subtypes and alterations of neutrophils in this process remain unclear. Here, we performed single-cell sequencing of cardiac CD45+ cells isolated from the murine myocardium subjected to MIRI at six-time points. We identified diverse types of infiltrating immune cells and their dynamic changes during MIRI. Cardiac neutrophils showed the most immediate response and largest changes and featured with functionally heterogeneous subpopulations, including Ccl3hi Neu and Ym-1hi Neu, which were increased at 6 h and 1 d after reperfusion, respectively. Ym-1hi Neu selectively expressed genes with protective effects and was, therefore, identified as a novel specific type of cardiac cell in the injured heart. Further analysis indicated that neutrophils and their subtypes orchestrated subsequent immune responses in the cardiac tissues, especially instructing the response of macrophages. The abundance of Ym-1hi Neu was closely correlated with the therapeutic efficacy of MIRI when neutrophils were specifically targeted by anti-Lymphocyte antigen 6 complex locus G6D (Ly6G) or anti-Intercellular cell adhesion molecule-1 (ICAM-1) neutralizing antibodies. In addition, a neutrophil subtype with the same phenotype as Ym-1hi Neu was detected in clinical samples and correlated with prognosis. Ym-1 inhibition exacerbated myocardial injury, whereas Ym-1 supplementation significantly ameliorated injury in MIRI mice, which was attributed to the tilt of Ym-1 on the polarization of macrophages toward the repair phenotype in myocardial tissue. Overall, our findings reveal the anti-inflammatory phenotype of Ym-1hi Neu and highlight its critical role in myocardial protection during the early stages of MIRI.
Collapse
Affiliation(s)
- Yalan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenyu Kang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zili Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongqiang Zhang
- Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanfei Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Xinxin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junyi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liangqingqing Yin
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xunxun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Ma
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Jiang
- Department of International Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Zhihao Lin
- Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Congzhu Ding
- Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Kim Yun Jin
- School of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang 43900, Malaysia
| | - Alexey Sarapultsev
- School of Medical Biology, South Ural State University, Chelyabinsk 620049, Russia
| | - Fangfei Li
- Shum Yiu Foon Sum Bik Chuen Memorial Centre for Cancer and Inflammation Research (CCIR), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ge Zhang
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
| | - Tian Xie
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Changjun Yin
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University, Munich 80336, Germany
| | - Xiang Cheng
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yue Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
21
|
Wang XX, Li ZH, Du HY, Liu WB, Zhang CJ, Xu X, Ke H, Peng R, Yang DG, Li JJ, Gao F. The role of foam cells in spinal cord injury: challenges and opportunities for intervention. Front Immunol 2024; 15:1368203. [PMID: 38545108 PMCID: PMC10965697 DOI: 10.3389/fimmu.2024.1368203] [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: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 04/17/2024] Open
Abstract
Spinal cord injury (SCI) results in a large amount of tissue cell debris in the lesion site, which interacts with various cytokines, including inflammatory factors, and the intrinsic glial environment of the central nervous system (CNS) to form an inhibitory microenvironment that impedes nerve regeneration. The efficient clearance of tissue debris is crucial for the resolution of the inhibitory microenvironment after SCI. Macrophages are the main cells responsible for tissue debris removal after SCI. However, the high lipid content in tissue debris and the dysregulation of lipid metabolism within macrophages lead to their transformation into foamy macrophages during the phagocytic process. This phenotypic shift is associated with a further pro-inflammatory polarization that may aggravate neurological deterioration and hamper nerve repair. In this review, we summarize the phenotype and metabolism of macrophages under inflammatory conditions, as well as the mechanisms and consequences of foam cell formation after SCI. Moreover, we discuss two strategies for foam cell modulation and several potential therapeutic targets that may enhance the treatment of SCI.
Collapse
Affiliation(s)
- Xiao-Xin Wang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Ze-Hui Li
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Hua-Yong Du
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Wu-Bo Liu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chun-Jia Zhang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Han Ke
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Run Peng
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - De-Gang Yang
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Jian-Jun Li
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Institute of Rehabilitation Medicine, China Rehabilitation Research Center, Beijing, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| |
Collapse
|
22
|
Duan J, Zhao Q, He Z, Tang S, Duan J, Xing W. Current understanding of macrophages in intracranial aneurysm: relevant etiological manifestations, signaling modulation and therapeutic strategies. Front Immunol 2024; 14:1320098. [PMID: 38259443 PMCID: PMC10800944 DOI: 10.3389/fimmu.2023.1320098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Macrophages activation and inflammatory response play crucial roles in intracranial aneurysm (IA) formation and progression. The outcome of ruptured IA is considerably poor, and the mechanisms that trigger IA progression and rupture remain to be clarified, thereby developing effective therapy to prevent subarachnoid hemorrhage (SAH) become difficult. Recently, climbing evidences have been expanding our understanding of the macrophages relevant IA pathogenesis, such as immune cells population, inflammatory activation, intra-/inter-cellular signaling transductions and drug administration responses. Crosstalk between macrophages disorder, inflammation and cellular signaling transduction aggravates the devastating consequences of IA. Illustrating the pros and cons mechanisms of macrophages in IA progression are expected to achieve more efficient treatment interventions. In this review, we summarized the current advanced knowledge of macrophages activation, infiltration, polarization and inflammatory responses in IA occurrence and development, as well as the most relevant NF-κB, signal transducer and activator of transcription 1 (STAT1) and Toll-Like Receptor 4 (TLR4) regulatory signaling modulation. The understanding of macrophages regulatory mechanisms is important for IA patients' clinical outcomes. Gaining insight into the macrophages regulation potentially contributes to more precise IA interventions and will also greatly facilitate the development of novel medical therapy.
Collapse
Affiliation(s)
- Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Qijie Zhao
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zeyuan He
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Shuang Tang
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Jia Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
23
|
Tawbeh A, Raas Q, Tahri-Joutey M, Keime C, Kaiser R, Trompier D, Nasser B, Bellanger E, Dessard M, Hamon Y, Benani A, Di Cara F, Cunha Alves T, Berger J, Weinhofer I, Mandard S, Cherkaoui-Malki M, Andreoletti P, Gondcaille C, Savary S. Immune response of BV-2 microglial cells is impacted by peroxisomal beta-oxidation. Front Mol Neurosci 2023; 16:1299314. [PMID: 38164407 PMCID: PMC10757945 DOI: 10.3389/fnmol.2023.1299314] [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: 09/26/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Microglia are crucial for brain homeostasis, and dysfunction of these cells is a key driver in most neurodegenerative diseases, including peroxisomal leukodystrophies. In X-linked adrenoleukodystrophy (X-ALD), a neuroinflammatory disorder, very long-chain fatty acid (VLCFA) accumulation due to impaired degradation within peroxisomes results in microglial defects, but the underlying mechanisms remain unclear. Using CRISPR/Cas9 gene editing of key genes in peroxisomal VLCFA breakdown (Abcd1, Abcd2, and Acox1), we recently established easily accessible microglial BV-2 cell models to study the impact of dysfunctional peroxisomal β-oxidation and revealed a disease-associated microglial-like signature in these cell lines. Transcriptomic analysis suggested consequences on the immune response. To clarify how impaired lipid degradation impacts the immune function of microglia, we here used RNA-sequencing and functional assays related to the immune response to compare wild-type and mutant BV-2 cell lines under basal conditions and upon pro-inflammatory lipopolysaccharide (LPS) activation. A majority of genes encoding proinflammatory cytokines, as well as genes involved in phagocytosis, antigen presentation, and co-stimulation of T lymphocytes, were found differentially overexpressed. The transcriptomic alterations were reflected by altered phagocytic capacity, inflammasome activation, increased release of inflammatory cytokines, including TNF, and upregulated response of T lymphocytes primed by mutant BV-2 cells presenting peptides. Together, the present study shows that peroxisomal β-oxidation defects resulting in lipid alterations, including VLCFA accumulation, directly reprogram the main cellular functions of microglia. The elucidation of this link between lipid metabolism and the immune response of microglia will help to better understand the pathogenesis of peroxisomal leukodystrophies.
Collapse
Affiliation(s)
- Ali Tawbeh
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Quentin Raas
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Mounia Tahri-Joutey
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences and Techniques, University Hassan I, Settat, Morocco
| | - Céline Keime
- Plateforme GenomEast, IGBMC, CNRS UMR 7104, Inserm U1258, University of Strasbourg, Illkirch, France
| | - Romain Kaiser
- Plateforme GenomEast, IGBMC, CNRS UMR 7104, Inserm U1258, University of Strasbourg, Illkirch, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| | - Boubker Nasser
- Laboratory of Biochemistry, Neurosciences, Natural Resources and Environment, Faculty of Sciences and Techniques, University Hassan I, Settat, Morocco
| | - Emma Bellanger
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Marie Dessard
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Yannick Hamon
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Francesca Di Cara
- Department of Microbiology and Immunology, Dalhousie University, IWK Health Centre, Halifax, NS, Canada
| | - Tânia Cunha Alves
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Stéphane Mandard
- LipSTIC LabEx, University of Bourgogne, INSERM LNC UMR1231, Dijon, France
| | | | | | | | - Stéphane Savary
- Laboratoire Bio-PeroxIL EA7270, University of Bourgogne, Dijon, France
| |
Collapse
|
24
|
Wang Q, Yi J, Liu H, Luo M, Yin G, Huang Z. Iguratimod promotes functional recovery after SCI by repairing endothelial cell tight junctions. Exp Neurol 2023; 368:114503. [PMID: 37572946 DOI: 10.1016/j.expneurol.2023.114503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Destruction of the blood-spinal cord barrier (BSCB) after spinal cord injury (SCI) is an important factor promoting the progression of the injury. This study addressed how to repair the BSCB in order to promote the repair of injured spinal cords. Iguratimod (IGU), an anti-rheumatic drug, has been approved for clinical use. A spinal cord injury mouse model and TNF-α-stimulated bEnd.3 cells were used to investigate the effect and mechanism of IGU on injured BSCB. An intracerebroventricular osmotic pump was used to administer drugs to the SCI mouse model. The results showed that the SCI mice in the treatment group had better recovery of neurological function than the control group. Examination of the tissue revealed better repair of the BSCB in injured spinal cords after medication. According to the results from the cell model, IGU promoted the expression of tight junction proteins and reduced cell permeability. Further research found that IGU repaired the barrier function by regulating glycolysis levels in the injured endothelial cells. In studying the mechanism, IGU was found to regulate HIF-1α expression through the NF-κB pathway, thereby regulating the expression of the glycolytic enzymes related to endothelial injury. In summary, IGU promoted functional recovery in vivo by repairing the BSCB. In vitro, IGU regulated the level of glycolysis in the damaged endothelium through the NF-κB pathway, thereby repairing the tight junctions between the endothelium. Therefore, IGU may become a potential drug for treating spinal cord injury.
Collapse
Affiliation(s)
- Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiang Yi
- Department of Orthopedics, Yancheng Third People's Hospital, Yancheng 224008, Jiangsu, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mingran Luo
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
25
|
Hamel R, Peruzzotti-Jametti L, Ridley K, Testa V, Yu B, Rowitch D, Marioni JC, Pluchino S. Time-resolved single-cell RNAseq profiling identifies a novel Fabp5+ subpopulation of inflammatory myeloid cells with delayed cytotoxic profile in chronic spinal cord injury. Heliyon 2023; 9:e18339. [PMID: 37636454 PMCID: PMC10450865 DOI: 10.1016/j.heliyon.2023.e18339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023] Open
Abstract
Traumatic spinal cord injuries (SCI) are a group of highly debilitating pathologies affecting thousands annually, and adversely affecting quality of life. Currently, no fully restorative therapies exist, and SCI still results in significant personal, societal and financial burdens. Inflammation plays a major role in the evolution of SCI, with myeloid cells, including bone marrow derived macrophages (BMDMs) and microglia (MG) being primary drivers of both early secondary pathogenesis and delayed wound healing events. The precise role of myeloid cell subsets is unclear as upon crossing the blood-spinal cord barrier, infiltrating bone marrow derived macrophages (BMDMs) may take on the morphology of resident microglia, and upregulate canonical microglia markers, thus making the two populations difficult to distinguish. Here, we used time-resolved scRNAseq and transgenic fate-mapping to chart the transcriptional profiles of tissue-resident and -infiltrating myeloid cells in a mouse model of thoracic contusion SCI. Our work identifies a novel subpopulation of foam cell-like inflammatory myeloid cells with increased expression of Fatty Acid Binding Protein 5 (Fabp5) and comprise both tissue-resident and -infiltrating cells. Fabp5+ inflammatory myeloid cells display a delayed cytotoxic profile that is predominant at the lesion epicentre and extends into the chronic phase of SCI.
Collapse
Affiliation(s)
- Regan Hamel
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | | | - Veronica Testa
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Bryan Yu
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - David Rowitch
- Cambridge Stem Cell Institute, University of Cambridge, UK
| | - John C. Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
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
|
27
|
Cao J, Pan C, Zhang J, Chen Q, Li T, He D, Cheng X. Analysis and verification of the circRNA regulatory network RNO_CIRCpedia_ 4214/RNO-miR-667-5p/Msr1 axis as a potential ceRNA promoting macrophage M2-like polarization in spinal cord injury. BMC Genomics 2023; 24:181. [PMID: 37020267 PMCID: PMC10077679 DOI: 10.1186/s12864-023-09273-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND CircRNAs are involved in the pathogenesis of several central nervous system diseases. However, their functions and mechanisms in spinal cord injury (SCI) are still unclear. Therefore, the purpose of this study was to evaluate circRNA and mRNA expression profiles in the pathological setting of SCI and to predict the potential function of circRNA through bioinformatics. METHODS A microarray-based approach was used for the simultaneous measurement of circRNAs and mRNAs, together with qPCR, fluorescence in situ hybridization, western immunoblotting, and dual-luciferase reporter assays to investigate the associated regulatory mechanisms in a rat SCI model. RESULTS SCI was found to be associated with the differential expression of 414 and 5337 circRNAs and mRNAs, respectively. Pathway enrichment analyses were used to predict the primary function of these circRNAs and mRNAs. GSEA analysis showed that differentially expressed mRNAs were primarily associated with inflammatory immune response activity. Further screening of these inflammation-associated genes was used to construct and analyze a competing endogenous RNA network. RNO_CIRCpedia_4214 was knocked down in vitro, resulting in reduced expression of Msr1, while the expression of RNO-miR-667-5p and Arg1 was increased. Dual-luciferase assays demonstrated that RNO_CIRCpedia_4214 bound to RNO-miR-667-5p. The RNO_CIRCpedia_4214/RNO-miR-667-5p/Msr1 axis may be a potential ceRNA that promotes macrophage M2-like polarization in SCI. CONCLUSION Overall, these results highlighted the critical role that circRNAs may play in the pathophysiology of SCI and the discovery of a potential ceRNA mechanism based on novel circRNAs that regulates macrophage polarization, providing new targets for the treatment of SCI.
Collapse
Affiliation(s)
- Jian Cao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Chongzhi Pan
- Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Jian Zhang
- Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi, 330006, China
| | - Qi Chen
- Jiangxi Key Laboratory of Intervertebral Disc Disease, Nanchang University, Jiangxi, 330006, China
| | - Tao Li
- Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Dingwen He
- Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi, 330006, China
| | - Xigao Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
- Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi, 330006, China.
- Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi, 330006, China.
- Jiangxi Key Laboratory of Intervertebral Disc Disease, Nanchang University, Jiangxi, 330006, China.
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, East Laker District, Nanchang, Jiangxi, China.
| |
Collapse
|
28
|
Jiang T, Qin T, Gao P, Tao Z, Wang X, Wu M, Gu J, Chu B, Zheng Z, Yi J, Xu T, Huang Y, Liu H, Zhao S, Ren Y, Chen J, Yin G. SIRT1 attenuates blood-spinal cord barrier disruption after spinal cord injury by deacetylating p66Shc. Redox Biol 2023; 60:102615. [PMID: 36716673 PMCID: PMC9900454 DOI: 10.1016/j.redox.2023.102615] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
Disruption of the blood-spinal cord barrier (BSCB) leads to inflammatory cell infiltration and neural cell death, thus, contributing to poor functional recovery after spinal cord injury (SCI). Previous studies have suggested that Sirtuin 1 (SIRT1), an NAD+-dependent class III histone deacetylase, is abundantly expressed in endothelial cells and promotes endothelial homeostasis. However, the role of SIRT1 in BSCB function after SCI remains poorly defined. Here, we report that SIRT1 is highly expressed in spinal cord endothelial cells, and its expression significantly decreases after SCI. Using endothelial cell-specific SIRT1 knockout mice, we observed that endothelial cell-specific knockout of SIRT1 aggravated BSCB disruption, thus, resulting in widespread inflammation, neural cell death and poor functional recovery after SCI. In contrast, activation of SIRT1 by the agonist SRT1720 had beneficial effects. In vitro, knockdown of SIRT1 exacerbated IL-1β-induced endothelial barrier disruption in bEnd.3 cells, whereas overexpression of SIRT1 was protective. Using RNA-seq and IP/MS analysis, we identified p66Shc, a redox protein, as the potential target of SIRT1. Further studies demonstrated that SIRT1 interacts with and deacetylates p66Shc, thereby attenuating oxidative stress and protecting endothelial barrier function. Overall, our results indicate that SIRT1 decreases endothelial ROS production and attenuates BSCB disruption by deacetylating p66Shc after SCI, and suggest that SIRT1 activation has potential as a therapeutic approach to promote functional recovery against BSCB disruption following SCI.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tao Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Peng Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhiwen Tao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xiaowei Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Mengyuan Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jun Gu
- Department of Orthopedics, Xishan People's Hospital, Wuxi, 214000, Jiangsu, China
| | - Bo Chu
- Department of Orthopedics, Xishan People's Hospital, Wuxi, 214000, Jiangsu, China
| | - Ziyang Zheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiang Yi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yifan Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Yongxin Ren
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
29
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [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: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
30
|
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 PMCID: PMC11415199 DOI: 10.1007/s10571-022-01203-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
31
|
Nance SA, Muir L, Delproprosto J, Lumeng CN. MSR1 is not required for obesity-associated inflammation and insulin resistance in mice. Sci Rep 2023; 13:2651. [PMID: 36788340 PMCID: PMC9927046 DOI: 10.1038/s41598-023-29736-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Obesity induces a chronic inflammatory state associated with changes in adipose tissue macrophages (ATMs). Macrophage scavenger receptor 1 (MSR1) has been implicated in the regulation of adipose tissue inflammation and diabetes pathogenesis; however, reports have been mixed on the contribution of MSR1 in obesity and glucose intolerance. We observed increased MSR1 expression in VAT of obese diabetic individuals compared to non-diabetic and single nuclear RNA sequencing identified macrophage-specific expression of MSR1 in human adipose tissue. We examined male Msr1-/- (Msr1KO) and WT controls and observed protection from obesity and AT inflammation in non-littermate Msr1KO mice. We then evaluated obese littermate Msr1+/- (Msr1HET) and Msr1KO mice. Both Msr1KO mice and Msr1HET mice became obese and insulin resistant when compared to their normal chow diet counterparts, but there was no Msr1-dependent difference in body weight, glucose metabolism, or insulin resistance. Flow cytometry revealed no significant differences between genotypes in ATM subtypes or proliferation in male and female mice. We observed increased frequency of proliferating ATMs in obese female compared to male mice. Overall, we conclude that while MSR1 is a biomarker of diabetes status in human adipose tissue, in mice Msr1 is not required for obesity-associated insulin resistance or ATM accumulation.
Collapse
Affiliation(s)
- Sierra A Nance
- Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI, 48109, USA
- Department of Pediatrics, University of Michigan Medical School, 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI, 48109, USA
| | - Lindsey Muir
- Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer Delproprosto
- Department of Pediatrics, University of Michigan Medical School, 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI, 48109, USA
| | - Carey N Lumeng
- Molecular and Integrative Physiology, University of Michigan Medical School, 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI, 48109, USA.
- Department of Pediatrics, University of Michigan Medical School, 109 Zina Pitcher Place, 2057 BSRB, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
32
|
Basu J, Madhulika S, Murmu KC, Mohanty S, Samal P, Das A, Mahapatra S, Saha S, Sinha I, Prasad P. Molecular and epigenetic alterations in normal and malignant myelopoiesis in human leukemia 60 (HL60) promyelocytic cell line model. Front Cell Dev Biol 2023; 11:1060537. [PMID: 36819104 PMCID: PMC9932920 DOI: 10.3389/fcell.2023.1060537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
In vitro cell line model systems are essential in supporting the research community due to their low cost, uniform culturing conditions, homogeneous biological resources, and easy experimental design to study the cause and effect of a gene or a molecule. Human leukemia 60 (HL60) is an in-vitro hematopoietic model system that has been used for decades to study normal myeloid differentiation and leukemia biology. Here, we show that IMDM supplemented with 20% FBS is an optimal culturing condition and induces effective myeloid differentiation compared with RPMI supplemented with 10% FBS when HL60 is induced with 1α,25-dihydroxyvitamin D3 (Vit D3) and all-trans retinoic acid (ATRA). The chromatin organization is compacted, and the repressive epigenetic mark H3K27me3 is enhanced upon HL60-mediated terminal differentiation. Differential gene expression analysis obtained from RNA sequencing in HL60 cells during myeloid differentiation showed the induction of pathways involved in epigenetic regulation, myeloid differentiation, and immune regulation. Using high-throughput transcriptomic data (GSE74246), we show the similarities (genes that did not satisfy |log2FC|>1 and FDR<0.05) and differences (FDR <0.05 and |log2FC|>1) between granulocyte-monocyte progenitor vs HL60 cells, Vit D3 induced monocytes (vMono) in HL60 cells vs primary monocytes (pMono), and HL60 cells vs leukemic blasts at the transcriptomic level. We found striking similarities in biological pathways between these comparisons, suggesting that the HL60 model system can be effectively used for studying myeloid differentiation and leukemic aberrations. The differences obtained could be attributed to the fact that the cellular programs of the leukemic cell line and primary cells are different. We validated several gene expression patterns for different comparisons with CD34+ cells derived from cord blood for myeloid differentiation and AML patients. In addition to the current knowledge, our study further reveals the significance of using HL60 cells as in vitro model system under optimal conditions to understand its potential as normal myeloid differentiation model as well as leukemic model at the molecular level.
Collapse
Affiliation(s)
- Jhinuk Basu
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,RCB, Regional Centre for Biotechnology, Faridabad, India
| | - Swati Madhulika
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,RCB, Regional Centre for Biotechnology, Faridabad, India
| | - Krushna Chandra Murmu
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,RCB, Regional Centre for Biotechnology, Faridabad, India
| | - Smrutishree Mohanty
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,RCB, Regional Centre for Biotechnology, Faridabad, India
| | - Priyanka Samal
- IMS and SUM Hospital, Siksha ‘O' Anusandhan University, Bhubaneswar, India
| | - Asima Das
- Department of Obstetrics and Gynecology, KIMS, Bhubaneswar, India
| | - Soumendu Mahapatra
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,Kalinga Institute of Industrial Technology (KIIT), School of Biotechnology, Bhubaneswar, India
| | - Subha Saha
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Indranil Sinha
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Solna, Sweden
| | - Punit Prasad
- Chromatin and Epigenetics Unit, Institute of Life Sciences, Bhubaneswar, India,*Correspondence: Punit Prasad,
| |
Collapse
|
33
|
Li C, Xiong W, Wan B, Kong G, Wang S, Wang Y, Fan J. Role of peripheral immune cells in spinal cord injury. Cell Mol Life Sci 2023; 80:2. [PMID: 36478290 PMCID: PMC9729325 DOI: 10.1007/s00018-022-04644-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/01/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Secondary spinal cord injury is caused by an inflammatory response cascade, and the process is irreversible. The immune system, as a mediator of inflammation, plays an important role in spinal cord injury. The spinal cord retains its immune privilege in a physiological state. Hence, elucidating the mechanisms by which peripheral immune cells are recruited to the lesion site and function after spinal cord injury is meaningful for the exploration of clinical therapeutic targets. In this review, we provide an overview of the multifaceted roles of peripheral immune cells in spinal cord injury.
Collapse
Affiliation(s)
- Cong Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wu Xiong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bowen Wan
- Department of Orthopaedics, Subei People's Hospital of Jiangsu, Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Guang Kong
- Nanjing Medical University, Nanjing, 210029, China
| | - Siming Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yingying Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
34
|
Sheng W, Ji G, Zhang L. Role of macrophage scavenger receptor MSR1 in the progression of non-alcoholic steatohepatitis. Front Immunol 2022; 13:1050984. [PMID: 36591228 PMCID: PMC9797536 DOI: 10.3389/fimmu.2022.1050984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD), and the dysregulation of lipid metabolism and oxidative stress are the typical features. Subsequent dyslipidemia and oxygen radical production may render the formation of modified lipids. Macrophage scavenger receptor 1 (MSR1) is responsible for the uptake of modified lipoprotein and is one of the key molecules in atherosclerosis. However, the unrestricted uptake of modified lipoproteins by MSR1 and the formation of cholesterol-rich foamy macrophages also can be observed in NASH patients and mouse models. In this review, we highlight the dysregulation of lipid metabolism and oxidative stress in NASH, the alteration of MSR1 expression in physiological and pathological conditions, the formation of modified lipoproteins, and the role of MSR1 on macrophage foaming and NASH development and progression.
Collapse
Affiliation(s)
| | | | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Wang X, Wen D, You C, Tao C, Ma L. Comprehensive analysis of immune cell infiltration and role of MSR1 expression in aneurysmal subarachnoid haemorrhage. Cell Prolif 2022:e13379. [PMID: 36515067 DOI: 10.1111/cpr.13379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Aneurysmal subarachnoid haemorrhage (aSAH), resulting from the rupture of intracranial aneurysms, can yield high mortality and disability. This study aimed to explore the immune infiltration of aneurysmal tissues and investigate a novel mechanism underlying aSAH. We downloaded datasets containing expression profiles of aneurysmal and normal arterial tissues from the online database. Then a comprehensive bioinformatic strategy was conducted to select the biomarkers of aneurysmal tissues. Two calculation algorithms were performed to identify the unique immune characteristics between aneurysmal tissues and normal arteries. Double immunofluorescence staining was used to investigate the role of pathway-related proteins in the inflammatory process after aSAH. Six microarray datasets were integrated, and another RNA-sequencing dataset was used as the validation dataset. Functional enrichment analysis of the differentially expressed genes indicated that immune-related processes were closely related to the progression of aSAH. We then performed immune microenvironment infiltration analysis, and the results suggested macrophages were abnormally enriched in aneurysmal tissues. Core gene MSR1 was filtered through a comprehensive bioinformatic strategy. Our analysis suggested that MSR1 might be associated with macrophage activation and migration. Our study elucidated the impact of macrophage and MSR1 on aSAH progression. These findings were helpful in gaining insight into the immune heterogeneity of aneurysmal tissues and normal arteries, and in identifying patients who might benefit from immunotherapy.
Collapse
Affiliation(s)
- Xing Wang
- Department of Neurosurgery, West China hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dingke Wen
- Department of Neurosurgery, West China hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China hospital, Sichuan University, Chengdu, Sichuan, China.,West China Brain Research Centre, Sichuan University, Chengdu, Sichuan, China
| | - Chuanyuan Tao
- Department of Neurosurgery, West China hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Ma
- Department of Neurosurgery, West China hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Feng Y, Peng Y, Jie J, Yang Y, Yang P. The immune microenvironment and tissue engineering strategies for spinal cord regeneration. Front Cell Neurosci 2022; 16:969002. [PMID: 35990891 PMCID: PMC9385973 DOI: 10.3389/fncel.2022.969002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regeneration of neural tissue is limited following spinal cord injury (SCI). Successful regeneration of injured nerves requires the intrinsic regenerative capability of the neurons and a suitable microenvironment. However, the local microenvironment is damaged, including insufficient intraneural vascularization, prolonged immune responses, overactive immune responses, dysregulated bioenergetic metabolism and terminated bioelectrical conduction. Among them, the immune microenvironment formed by immune cells and cytokines plays a dual role in inflammation and regeneration. Few studies have focused on the role of the immune microenvironment in spinal cord regeneration. Here, we summarize those findings involving various immune cells (neutrophils, monocytes, microglia and T lymphocytes) after SCI. The pathological changes that occur in the local microenvironment and the function of immune cells are described. We also summarize and discuss the current strategies for treating SCI with tissue-engineered biomaterials from the perspective of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jing Jie
- Department of Clinical Laboratory, The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong, China
- Jing Jie,
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Yumin Yang,
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin Medical University, Harbin, China
- *Correspondence: Pengxiang Yang,
| |
Collapse
|
37
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
38
|
Xia M, Zhang Q, Zhang Y, Li R, Zhao T, Chen L, Liu Q, Zheng S, Li H, Qian Z, Yang L. Growth Differentiation Factor 15 Regulates Oxidative Stress-Dependent Ferroptosis Post Spinal Cord Injury by Stabilizing the p62-Keap1-Nrf2 Signaling Pathway. Front Aging Neurosci 2022; 14:905115. [PMID: 35860670 PMCID: PMC9289442 DOI: 10.3389/fnagi.2022.905115] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/31/2022] [Indexed: 01/12/2023] Open
Abstract
Background Spinal cord injury (SCI) is a severe traumatic disorder of the central nervous system (CNS) that causes irreversible damage to the nervous tissue. The consequent hemorrhage contributed by trauma induces neuronal ferroptosis post SCI, which is an important death mode to mediate neuronal loss. Growth differentiation factor 15 (GDF15) is a cytokine that regulates cell proliferation, differentiation, and death. However, the specific role of GDF15 in neuronal ferroptosis post SCI remains unknown. Materials and Methods Neuronal ferroptosis in vitro was measured by detection of lipid peroxidation, glutathione, iron content, and reactive oxidative stress. In vivo, western blotting and immunofluorescence (IF) staining was utilized to measure ferroptosis post SCI. IF staining, TUNEL staining, hematoxylin-eosin staining, and Nissl staining were used to measure neurological damage. Finally, locomotor function recovery was analyzed using the Basso Mouse Scale and Louisville Swim Scale. Results GDF15 was significantly increased in neuronal ferroptosis and silencing GDF15 aggravated ferroptosis both in vitro and in vivo. Besides, GDF15-mediated inhibition of neuronal ferroptosis is through p62-dependent Keap1-Nrf2 pathway. In SCI mice, knockdown of GDF15 significantly exacerbated neuronal death, interfered with axon regeneration and remyelination, aggravated ferroptosis-mediated neuroinflammation, and restrained locomotor recovery. Conclusion GDF15 effectively alleviated neuronal ferroptosis post SCI via the p62-Keap1-Nrf2 signaling pathway and promoted locomotor recovery of SCI mice, which is suggested as a potential target on SCI pathogenesis and treatment.
Collapse
Affiliation(s)
- Mingjie Xia
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qinyang Zhang
- Postgraduate School, Dalian Medical University, Dalian, China
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
| | - Yanan Zhang
- Postgraduate School, Dalian Medical University, Dalian, China
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
| | - Rulin Li
- Postgraduate School, Dalian Medical University, Dalian, China
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
| | - Tianyu Zhao
- Postgraduate School, Dalian Medical University, Dalian, China
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
| | - Lingxia Chen
- Department of Cardiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiangxian Liu
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shengnai Zheng
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Haijun Li
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
- Taizhou Clinical Medical School of Nanjing Medical University, Taizhou, China
- *Correspondence: Haijun Li,
| | - Zhanyang Qian
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, China
- School of Medicine, Southeast University, Nanjing, China
- Zhanyang Qian,
| | - Lei Yang
- Department of Orthopedics, Taizhou People’s Hospital, Nanjing Medical University, Taizhou, China
- Taizhou Clinical Medical School of Nanjing Medical University, Taizhou, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Lei Yang,
| |
Collapse
|
39
|
Xie Y, Jia Y, Li Z, Hu F. Scavenger receptor A in immunity and autoimmune diseases: Compelling evidence for targeted therapy. Expert Opin Ther Targets 2022; 26:461-477. [PMID: 35510370 DOI: 10.1080/14728222.2022.2072729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Scavenger receptor A (SR-A) is reported to be involved in innate and adaptive immunity and in recent years, the soluble form of SR-A has also been identified. Intriguingly, SR-A displays double-edged sword features in different diseases. Moreover, targeted therapy on SR-A, including genetic modulation, small molecule inhibitor, inhibitory peptides, fucoidan, and blocking antibodies, provides potential strategies for treatment. Currently, therapeutics targeting SR-A are in preclinical studies and clinical trials, revealing great perspectives in future immunotherapy. AREAS COVERED Through searching PubMed (January 1979-March 2022) and clinicaltrials.gov, we review most of the research and clinical trials involving SR-A. This review briefly summarizes recent study advances on SR-A, with particular concern on its role in immunity and autoimmune diseases. EXPERT OPINION Given the emerging evidence of SR-A in immunity, its targeted therapy has been studied in various diseases, especially autoimmune diseases. However, many challenges still remain to be overcome, such as the double-sworded effects and the specific isoform targeting. For further clinical success of SR-A targeted therapy, the crystal structure illustration and the dual function discrimination of SR-A should be further investigated. Nevertheless, although challenging, targeting SR-A would be a potential effective strategy in the treatment of autoimmune diseases and other immune-related diseases.
Collapse
Affiliation(s)
- Yang Xie
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, Peking, China
| | - Yuan Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, Peking, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, Peking, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Peking, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, Peking, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, Peking, China.,State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, Peking, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, Peking, China
| |
Collapse
|
40
|
Miron J, Picard C, Labonté A, Auld D, Poirier J. MSR1 and NEP Are Correlated with Alzheimer's Disease Amyloid Pathology and Apolipoprotein Alterations. J Alzheimers Dis 2022; 86:283-296. [PMID: 35034907 DOI: 10.3233/jad-215410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In mouse models of amyloidosis, macrophage receptor 1 (MSR1) and neprilysin (NEP) have been shown to interact to reduce amyloid burden in the brain. OBJECTIVE The purpose of this study is to analyze these two gene products in combination with apolipoproteins and Aβ1-42 in the cerebrospinal fluid (CSF) and plasma of individuals at different stages of Alzheimer's disease (AD), as well as in autopsied brain samples from ROSMAP (Religious Orders Study and Memory and Aging Project). METHODS CSF/plasma levels of MSR1 and NEP were measured using the sensitive primer extension assay technology. CSF Aβ1-42 was assessed with ELISA, while CSF ApoE and ApoJ were measured with the Luminex's multiplex technology. Brain MSR1, APOE, and CLU (APOJ) mRNA levels were measured with RNA-Seq and contrasted to amyloid plaques pathology using CERAD staging. RESULTS While plasma and CSF MSR1 levels are significantly correlated, this correlation was not observed for NEP. In addition to be highly correlated to one another, CSF levels of both MSR1 and NEP are strongly correlated with AD status and CSF Aβ1-42, ApoE, and ApoJ levels. In the cortical tissues of subjects from ROSMAP, MSR1 mRNA levels are correlated with CLU mRNA levels and the CERAD scores but not with APOE mRNA levels. CONCLUSION The discrepancies observed between CSF/plasma levels of MSR1 and NEP with CSF Aβ1-42 and ApoE concentrations can be explained by many factors, such as the disease stage or the involvement of the blood-brain barrier breakdown that leads to the infiltration of peripheral monocytes or macrophages.
Collapse
Affiliation(s)
- Justin Miron
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada.,McGill University, Montréal, QC, Canada
| | - Cynthia Picard
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Anne Labonté
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | | | - Judes Poirier
- Douglas Hospital Research Centre, Montréal, QC, Canada.,Centre for the Studies on the Prevention of Alzheimer's Disease, Montréal, QC, Canada.,McGill University, Montréal, QC, Canada
| | | |
Collapse
|
41
|
Wang B, Tang X, Yao L, Wang Y, Chen Z, Li M, Wu N, Wu D, Dai X, Jiang H, Ai D. Disruption of USP9X in macrophages promotes foam cell formation and atherosclerosis. J Clin Invest 2022; 132:154217. [PMID: 35389885 PMCID: PMC9106359 DOI: 10.1172/jci154217] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
Subendothelial macrophage internalization of modified lipids and foam cell formation are hallmarks of atherosclerosis. Deubiquitinating enzymes (DUBs) are involved in various cellular activities; however, their role in foam cell formation is not fully understood. Here, using a loss-of-function lipid accumulation screening, we identified ubiquitin-specific peptidase 9 X-linked (USP9X) as a factor that suppressed lipid uptake in macrophages. We found that USP9X expression in lesional macrophages was reduced during atherosclerosis development in both humans and rodents. Atherosclerotic lesions from macrophage USP9X-deficient mice showed increased macrophage infiltration, lipid deposition, and necrotic core content than control apolipoprotein E–KO (Apoe–/–) mice. Additionally, loss-of-function USP9X exacerbated lipid uptake, foam cell formation, and inflammatory responses in macrophages. Mechanistically, the class A1 scavenger receptor (SR-A1) was identified as a USP9X substrate that removed the K63 polyubiquitin chain at the K27 site. Genetic or pharmacological inhibition of USP9X increased SR-A1 cell surface internalization after binding of oxidized LDL (ox-LDL). The K27R mutation of SR-A1 dramatically attenuated basal and USP9X knockdown–induced ox-LDL uptake. Moreover, blocking binding of USP9X to SR-A1 with a cell-penetrating peptide exacerbated foam cell formation and atherosclerosis. In this study, we identified macrophage USP9X as a beneficial regulator of atherosclerosis and revealed the specific mechanisms for the development of potential therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Biqing Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xuening Tang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Liu Yao
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yuxin Wang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhipeng Chen
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Mengqi Li
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Naishi Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Dawei Wu
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangchen Dai
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongfeng Jiang
- Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ding Ai
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
42
|
Erythropoietin promotes M2 macrophage phagocytosis of Schwann cells in peripheral nerve injury. Cell Death Dis 2022; 13:245. [PMID: 35296651 PMCID: PMC8927417 DOI: 10.1038/s41419-022-04671-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022]
Abstract
Following acute sciatic nerve crush injury (SNCI), inflammation and the improper phagocytic clearance of dying Schwann cells (SCs) has effects on remodeling that lead to morbidity and incomplete functional recovery. Therapeutic strategies like the use of erythropoietin (EPO) for peripheral nerve trauma may serve to bring immune cell phagocytotic clearance under control to support debris clearance. We evaluated EPO’s effect on SNCI and found EPO treatment increased myelination and sciatic functional index (SFI) and bolstered anti-apoptosis and phagocytosis of myelin debris via CD206+ macrophages when compared to saline treatment. EPO enhanced M2 phenotype activity, both in bone marrow-derived macrophages (BMMØs) and peritoneal-derived macrophages (PMØs) in vitro, as well as in PMØs in vivo. EPO increased efferocytosis of apoptotic sciatic nerve derived Schwann cells (SNSCs) in both settings as demonstrated using immunofluorescence (IF) and flow cytometry. EPO treatment significantly attenuated pro-inflammatory genes (IL1β, iNOS, and CD68) and augmented anti-inflammatory genes (IL10 and CD163) and the cell-surface marker CD206. EPO also increased anti-apoptotic (Annexin V/7AAD) effects after lipopolysaccharide (LPS) induction in macrophages. Our data demonstrate EPO promotes the M2 phenotype macrophages to ameliorate apoptosis and efferocytosis of dying SCs and myelin debris and improves SN functional recovery following SNCI.
Collapse
|
43
|
Zhou Z, Li C, Bao T, Zhao X, Xiong W, Luo C, Yin G, Fan J. Exosome-shuttled miR-672-5p from anti-inflammatory microglia repair traumatic spinal cord injury by inhibiting AIM2/ASC/Caspase-1 signaling pathway mediated neuronal pyroptosis. J Neurotrauma 2022; 39:1057-1074. [PMID: 35243913 DOI: 10.1089/neu.2021.0464] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traumatic spinal cord injury (TSCI) is a devastating traumatic disease of the central nervous system, which leads to refractory loss of motor and sensory function. So far, there is no effective treatment for TSCI. Recently, however, nano-sized exosomes from various spinal cord cells have shown great prospects in the treatment of various diseases, including TSCI. Microglia are one of the components of the spinal cord microenvironment. Anti-inflammatory microglia (M2) have been shown to inhibit inflammation and promote the functional recovery of spinal cord after TSCI. However, the role micro RNAs (miRNAs) in exosomes derived from M2 microglia in the treatment of TSCI is unclear. In this study, we investigated whether M2 microglial exosomes (M2-Exos) could better promote the functional behavior recovery of mice with TSCI than M0 microglial exosomes (Exos). Compared with Exos, M2-Exos were found to have a better effect in promoting the recovery of functional behavior, promoting axon regeneration and reducing the level of pyroptosis of spinal cord neurons after TSCI. Through a series of experiments, we also confirmed that miR-672-5p is the most critical miRNA associated with M2-Exos, and that its targeting gene is AIM2. M2-Exos rich in miR-672-5p could inhibit the AIM2/ASC/Caspase-1 signaling pathway by inhibiting AIM2 activity, so as to inhibit neuronal pyroptosis and finally promote the recovery of functional behavior in mice with TSCI. In conclusion, our study suggests that the application of M2-Exos may be a promising treatment strategy for TSCI.
Collapse
Affiliation(s)
- Zheng Zhou
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Emergency Medicine, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, China, Nanjing, China, 210029;
| | - Cong Li
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| | - Tianyi Bao
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| | - Xuan Zhao
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| | - Wu Xiong
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| | - Chunyang Luo
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Emergency Medicine, Nanjing, Jiangsu, China;
| | - Guoyong Yin
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| | - Jin Fan
- Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 74734, Department of Orthopaedics, Nanjing, Jiangsu, China;
| |
Collapse
|
44
|
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: 19] [Impact Index Per Article: 6.3] [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
|
45
|
Wang B, Chang M, Zhang R, Wo J, Wu B, Zhang H, Zhou Z, Li Z, Zhang F, Zhong C, Tang S, Yang S, Sun G. Spinal cord injury target-immunotherapy with TNF-α autoregulated and feedback-controlled human umbilical cord mesenchymal stem cell derived exosomes remodelled by CRISPR/Cas9 plasmid. BIOMATERIALS ADVANCES 2022; 133:112624. [PMID: 35525736 DOI: 10.1016/j.msec.2021.112624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/26/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022]
Abstract
Human umbilical cord mesenchymal stem cell (hucMSC) derived exosomes (EXOs) have been investigated as a new treatment for spinal cord injury (SCI) because of their anti-inflammatory, anti-apoptotic, angiogenesis-promoting, and axonal regeneration properties. The CAQK peptide found in the brains of mice and humans after trauma has recently been found to specifically bind to the injured site after SCI. Thus, we developed a nanocarrier system called EXO-C@P based on hucMSC exosomes remodelled by the CRISPR/Cas9 plasmid to control inflammation and modified by the CAQK peptide. EXO-C@P was shown to effectively accumulate at the injury site and saturate the macrophages to significantly reduce the expression of inflammatory cytokines in a mouse model of SCI. Moreover, EXO-C@P treatment improved the performance of mice in behavioural assessments and upregulated soluble tumour necrosis factor receptor-1 (sTNFR1) in serum and at the trauma site after SCI surgery, but lowered the proportion of iNOS+ cells and the concentration of proinflammatory factors. In conclusion, EXO-C@P provides an effective alternative to multiple topical administration and drug delivery approaches for the treatment of SCI. STATEMENT OF SIGNIFICANCE: SCI is a serious disease characterised by a high incidence, high disability rate, and high medical costs, and has become a global medical problem. Several studies have shown that the inflammatory response is the critical inducer of secondary injury after SCI. The inflammatory cytokine TNF-α is considered to be one of the most significant therapeutic targets for autoimmune diseases. Antibodies targeting TNF-α and sTNFR1 are capable of neutralising free TNF-α. In this study, exosomes in the CRISPR/Cas9 system were used to establish stem cells with an autoregulated and feedback-controlled TNF-α response, with these cells secreting sTNFR1, which neutralised TNF-α and antagonised the inflammation stimulated by TNF-α. Moreover, the plasmid was combined with CAQK, which targeted the injury site and promoted the recovery of SCI function.
Collapse
Affiliation(s)
- Baocheng Wang
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Minmin Chang
- School of Chinese Medicine, Jinan University, Guangzhou, Guangdong Province 510632, China
| | - Renwen Zhang
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; School of Chinese Medicine, Jinan University, Guangzhou, Guangdong Province 510632, China
| | - Jin Wo
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, China
| | - Bowen Wu
- Department of Orthopedics, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU, Zhuzhou 412007, China
| | - Hua Zhang
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, Guangdong, China; The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou 510632, China
| | - Zhigang Zhou
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Zhizhong Li
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Feng Zhang
- Intensive Care Unit, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Cheng Zhong
- The Affiliated Hospital (Jiangmen Traditional Chinese Medicine Hospital), Jinan University, Guangzhou 510632, China
| | - Shujie Tang
- School of Chinese Medicine, Jinan University, Guangzhou, Guangdong Province 510632, China.
| | - Shuxian Yang
- Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, China; The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou 510632, China.
| | - Guodong Sun
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan 517000, China; Department of Orthopedics, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
46
|
Myelin and non-myelin debris contribute to foamy macrophage formation after spinal cord injury. Neurobiol Dis 2022; 163:105608. [PMID: 34979258 PMCID: PMC8783370 DOI: 10.1016/j.nbd.2021.105608] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 02/03/2023] Open
Abstract
Tissue damage after spinal cord injury (SCI) elicits a robust inflammatory cascade that fails to resolve in a timely manner, resulting in impaired wound healing and cellular regeneration. This inflammatory response is partly mediated by infiltrating immune cells, including macrophages. As professional phagocytes, macrophages initially play an important role in debris clearance at the injury site, which would be necessary for proper tissue regeneration. After SCI, most macrophages become filled with lipid droplets due to excessive uptake of lipid debris, assuming a "foamy" phenotype that is associated with a proinflammatory state. Myelin has been assumed to be the main source of lipid that induces foamy macrophage formation after injury given its abundance in the spinal cord. This assumption has led to the widespread use of purified myelin treatment to model foamy macrophage formation in vitro. However, the assumption that myelin is necessary for foamy macrophage formation remains untested. To this end, we developed a novel foamy macrophage assay utilizing total spinal cord homogenate to include all sources of lipid present at the injury site. Using the myelin basic protein knockout (MBP KO, i.e., Shiverer) mice that lack myelin, we investigated lipid accumulation in foamy macrophages. Primary macrophages treated with myelin-deficient spinal cord homogenate still formed large lipid droplets typically observed in foamy macrophages, although to a lesser degree than cells treated with normal homogenate. Similarly, MBP KO mice subjected to contusive spinal cord injury also formed foamy macrophages that exhibited reduced lipid content and associated with improved histological outcomes and reduced immune cell infiltration. Therefore, the absence of myelin does not preclude foamy macrophage formation, indicating that myelin is not the only major source of lipid that contributes this pathology, even though myelin may alter certain aspects of its inflammatory profile.
Collapse
|
47
|
Lin J, Shi J, Min X, Chen S, Zhao Y, Zhang Y, Cheng L. The GDF11 Promotes Nerve Regeneration After Sciatic Nerve Injury in Adult Rats by Promoting Axon Growth and Inhibiting Neuronal Apoptosis. Front Bioeng Biotechnol 2022; 9:803052. [PMID: 35059389 PMCID: PMC8764262 DOI: 10.3389/fbioe.2021.803052] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 11/20/2022] Open
Abstract
Introduction: Sciatic nerve injury is a common injury of the nervous system. Stem cell-based therapies, drug-based therapies and rehabilitation physiotherapy therapies are currently available, but their limited therapeutic efficacy limits their use. Here, we aimed to explore a novel lentiviral-based gene therapeutic strategy and to elaborate its mechanism. Materials and Methods: Recombinant GDF11 protein was used for the in vitro treatment of dorsal root ganglion (DRG) cells. Lentivirus was used to construct a vector system for the in vivo expression of GDF11. The nerve conduction function was detected using action-evoked potentials at different time periods, and the regulatory effect of nerves on target organs was detected by weighing the gastrocnemius muscle. Immunofluorescence of NF200 and S100 was used to show the regeneration of the sciatic nerve, and myelin and Nissl staining were performed to observe the pathological features of the tissue. Western was used to validate signaling pathways. The expression of related genes was observed by qPCR and Western blotting, and cell apoptosis was detected by flow cytometry. Result: GDF11 promotes the axonal growth of DRG cells and inhibits DGR cell apoptosis in vitro. GDF11 acts by activating the Smad pathway. GDF11 promotes the recovery of damaged sciatic nerve function in rats, the regeneration of damaged sciatic nerves in rats, and myelin regeneration of damaged sciatic nerves in rats. GDF11 also exerts a protective effect on neuronal cells in rats. Conclusion: Based on the present study, we conclude that GDF11 promotes axonal growth and inhibits DRG cell apoptosis in vitro through the Smad pathway, and lentivirus-mediated GDF11 overexpression in vivo can promote the recovery of sciatic nerves after transection by promoting axonal growth and inhibiting neuronal apoptosis in the spinal cord.
Collapse
Affiliation(s)
- Junhao Lin
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Shi
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cheeloo College of Medicine, Shandong University, Jinan, China.,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Min
- Department of Health Management Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Si Chen
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yunpeng Zhao
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanqiang Zhang
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Cheng
- Department of Orthopaedic, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
48
|
Li F, Sun X, Zheng B, Sun K, Zhu J, Ji C, Lin F, Huan L, Luo X, Yan C, Xu J, Hong Y, Wang Y, Xu X, Sun J, Song Z, Kong F, Shi J. Arginase II Promotes Intervertebral Disc Degeneration Through Exacerbating Senescence and Apoptosis Caused by Oxidative Stress and Inflammation via the NF-κB Pathway. Front Cell Dev Biol 2021; 9:737809. [PMID: 34926442 PMCID: PMC8679914 DOI: 10.3389/fcell.2021.737809] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Intervertebral disc degeneration (IDD) has been generally accepted as the major cause of low back pain (LBP), which imposes massive clinical and socioeconomic burdens. Previous studies have demonstrated that oxidative stress and inflammation-induced senescence and apoptosis of nucleus pulposus cells (NPCs) are the main cellular processes that cause IDD. Arginase II (ARG2), an enzyme involved in a variety of pathological processes, including cellular senescence, apoptosis, oxidative stress, and inflammation, has been shown to promote degeneration in several degenerative diseases, including osteoarticular diseases. Based on previous studies, we hypothesized that ARG2 deficiency might be conducive to the treatment of IDD by inhibiting the dyshomeostasis of the extracellular matrix (ECM), and the oxidative stress and inflammatory response-induced senescence and apoptosis via NF-κB. In this study, we found that ARG2 deficiency inhibited senescence and apoptosis of NPCs, and degeneration of the ECM induced by oxidative stress and the inflammatory response. Similar results were found with the selective NF-κB pathway inhibitor JSH-23. In contrast, overexpression of ARG2 had the opposite effect. Taken together, our results suggest that ARG2 deficiency prevents IDD via NF-κB, and may therefore, be a potential therapeutic strategy for IDD.
Collapse
Affiliation(s)
- Fudong Li
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiaofei Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bing Zheng
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kaiqiang Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chenglong Ji
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Feng Lin
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Le Huan
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xi Luo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen Yan
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiashun Xu
- The 905th Hospital of the People's Liberation Army Navy of China, Shanghai, China
| | - Yun Hong
- The 905th Hospital of the People's Liberation Army Navy of China, Shanghai, China
| | - Yuan Wang
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ximing Xu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zheming Song
- The 905th Hospital of the People's Liberation Army Navy of China, Shanghai, China
| | - Fanqi Kong
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiangang Shi
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
49
|
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: 15] [Impact Index Per Article: 3.8] [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
|
50
|
Dong L, Cao Y, Hou Y, Liu G. N 6 -methyladenosine RNA methylation: A novel regulator of the development and function of immune cells. J Cell Physiol 2021; 237:329-345. [PMID: 34515345 DOI: 10.1002/jcp.30576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022]
Abstract
N6 -methyladenosine (m6 A) RNA methylation is a reversible posttranscriptional modification in eukaryotes involving three types of functional proteins: "writers", "erasers", and "readers". m6 A regulates the metabolism of messenger RNAs and noncoding RNAs through RNA structure, splicing, stability, export, and translation, thereby participating in various physiological and pathological processes. Here, we summarize the current state of m6 A methylation researches, focusing on how these modifications modulate the fate decisions of innate and adaptive immune cells and regulate immune responses in immune-associated diseases, including viral infections and cancer. These studies showed that m6 A modifications and m6 A modifying proteins play a critical role in pathogen recognition, immune cell activation, immune cell fate decisions, and immune reactions. m6 A is a novel regulator of immune system homeostasis and activation.
Collapse
Affiliation(s)
- Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|