1
|
Wang X, Niu X, Wang Y, Liu Y, Yang C, Chen X, Qi Z. C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 pathway as a therapeutic target and regulatory mechanism for spinal cord injury. Neural Regen Res 2025; 20:2231-2244. [PMID: 39104168 DOI: 10.4103/nrr.nrr-d-24-00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/07/2024] [Indexed: 08/07/2024] Open
Abstract
Spinal cord injury involves non-reversible damage to the central nervous system that is characterized by limited regenerative capacity and secondary inflammatory damage. The expression of the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis exhibits significant differences before and after injury. Recent studies have revealed that the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis is closely associated with secondary inflammatory responses and the recruitment of immune cells following spinal cord injury, suggesting that this axis is a novel target and regulatory control point for treatment. This review comprehensively examines the therapeutic strategies targeting the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis, along with the regenerative and repair mechanisms linking the axis to spinal cord injury. Additionally, we summarize the upstream and downstream inflammatory signaling pathways associated with spinal cord injury and the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis. This review primarily elaborates on therapeutic strategies that target the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis and the latest progress of research on antagonistic drugs, along with the approaches used to exploit new therapeutic targets within the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis and the development of targeted drugs. Nevertheless, there are presently no clinical studies relating to spinal cord injury that are focusing on the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis. This review aims to provide new ideas and therapeutic strategies for the future treatment of spinal cord injury.
Collapse
Affiliation(s)
- Xiangzi Wang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaofei Niu
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingkai Wang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yang Liu
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Cheng Yang
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xuyi Chen
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Zhongquan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
- Fujian Maternity and Child Health Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Lv M, Zhao Y, Chang S, Gao Z. Identifying signature genes and their associations with immune cell infiltration in spinal cord injury. IBRO Neurosci Rep 2024; 17:320-328. [PMID: 39430218 PMCID: PMC11490871 DOI: 10.1016/j.ibneur.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
Background Early detection of spinal cord injury (SCI) is conducive to improving patient outcomes. In addition, many studies have revealed the role of immune cells in the progression or treatment of SCI. The objective of this study was to identify the early signature genes and clarify how they are related to immune cell infiltration in SCI. Methods We analysed and identified early signature genes associated with SCI via bioinformatics analysis of the GSE151371 dataset from the GEO database. These genes were subsequently verified in the GSE33886 dataset and qRT-PCR. Finally, the CIBERSORT algorithm was used to examine the immune cell infiltration in SCI and its relationship with signature genes. Results Seven SCI-related signature genes, including ARG1, RETN, BPI, GGH, CCNB1, HIST1H2AC, and HIST1H2BJ, were identified, and their expression was verified via an external validation cohort and qRT-PCR. Moreover, the ROC curves revealed the diagnostic value of these genes. In addition, on the basis of immune cell infiltration analysis, plasma cells, M0 macrophages, activated CD4+ memory T cells, γδ T cells, naive CD4+ T cells, and resting CD4+ memory T cells may participate in the progression of SCI. Conclusion This study identified seven early signature genes of SCI that may serve as biomarkers for the early diagnosis of SCI and contribute to our understanding of immune changes during the pathology of SCI.
Collapse
Affiliation(s)
- Meng Lv
- Department of Orthopaedics, Shaanxi Provincial People's Hospital (Third Affiliated Hospital of Xi’an Jiaotong University), Xi’an, Shaanxi Province 710068, China
| | - Yingjie Zhao
- Department of Orthopaedics, Shaanxi Provincial People's Hospital (Third Affiliated Hospital of Xi’an Jiaotong University), Xi’an, Shaanxi Province 710068, China
| | - Su’e Chang
- Department of Orthoapedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Zhengchao Gao
- Department of Orthoapedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| |
Collapse
|
3
|
Zhu L, Wang F, Xing J, Hu X, Gou X, Li J, Pang R, Zhang A. Modulatory effects of gut microbiota on innate and adaptive immune responses following spinal cord injury. Exp Neurol 2024; 379:114866. [PMID: 38876194 DOI: 10.1016/j.expneurol.2024.114866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/18/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Spinal cord injury (SCI) represents a highly debilitating trauma to the central nervous system, currently lacking effective therapeutic strategies. The cascade of inflammatory responses induced by secondary damage following SCI disrupts the local immune environment at the injury site, ultimately exacerbating functional impairments post-injury. With advancing research on the gut-brain axis, evidence suggests that dysbiosis of the gut microbiota post-SCI amplifies inflammatory responses and plays a pivotal role in modulating post-injury immune-inflammatory responses. In this review article, we will explore the significant role of the gut microbiota and its metabolic products in modulating the responses of central and peripheral immune cells post-SCI, as well as their potential as therapeutic interventions for SCI treatment.
Collapse
Affiliation(s)
- Li Zhu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Fangfang Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jiajia Xing
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Xiaomin Hu
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Xiang Gou
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Jiayu Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China.
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Gonzalez CE, Vaidya RS, Clayton SW, Tang SY. Secreted chemokines reveal diverse inflammatory and degenerative processes in the intervertebral disc of the STZ-HFD mouse model of Type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605332. [PMID: 39131361 PMCID: PMC11312574 DOI: 10.1101/2024.07.31.605332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The chronic inflammation present in type 2 diabetes causes many chronic inflammatory comorbidities, including cardiovascular, renal, and neuropathic complications. Type 2 diabetes is also associated with a number of spinal pathologies, including intervertebral disc (IVD) degeneration and chronic neck and back pain. Although confounding factors such as obesity are thought to increase the loads to the musculoskeletal system and subsequent degeneration, studies have shown that even after adjusting age, body mass index, and genetics (e.g. twins), patients with diabetes suffer from disproportionately more IVD degeneration and back pain. Yet the tissue-specific responses of the IVD during diabetes remains relatively unknown. We hypothesize that chronic diabetes fosters a proinflammatory microenvironment within the IVD that accelerates degeneration and increases susceptibility to painful disorders. To test this hypothesis, we evaluated two commonly used mouse models of diabetes - the leptin-receptor deficient mouse (db/db) and the chronic high-fat diet in mice with impaired beta-cell function (STZ-HFD). The db/db is a genetic model that spontaneous develop diabetes through hyperphagia, while the STZ-HFD mouse first exhibits rapid obesity development under HFD and pronounced insulin resistance following streptozotocin administration. Both animal models were allowed to develop sustained diabetes for at least twelve weeks, as defined by elevated hemoglobin A1C, hyperglycemia, and glucose intolerance. Following the twelve-week period, the IVDs were extracted in quantified in several measures including tissue-specific secreted cytokines, viscoelastic mechanical behavior, structural composition, and histopathologic degeneration. Although there were no differences in mechanical function or the overall structure of the IVD, the STZ-HFD IVDs were more degenerated. More notably, the STZ-HFD model shows a significantly higher fold increase for eight cytokines: CXCL2, CCL2, CCL3, CCL4, CCL12 (monocyte/macrophage associated), IL-2, CXCL9 (T-cell associated), and CCL5 (pleiotropic). Correlative network analyses revealed that the expression of cytokines differentially regulated between the db/db and the STZ-HFD models. Moreover, the STZ-HFD contained a fragmented and modular cytokine network, indicating greater complexities in the regulatory network. Taken together, the STZ-HFD model of type 2 diabetes may better recapitulate the complexities of the chronic inflammatory processes in the IVD during diabetes.
Collapse
Affiliation(s)
- Christian E. Gonzalez
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
| | - Rachana S. Vaidya
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Sade W. Clayton
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| | - Simon Y. Tang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO
- Institute of Material Science and Engineering, Washington University in St. Louis, St. Louis, MO
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
5
|
Lv J, Wang Z, Wang B, Deng C, Wang W, Sun L. S100A9 Induces Macrophage M2 Polarization and Immunomodulatory Role in the Lesion Site After Spinal Cord Injury in Rats. Mol Neurobiol 2024; 61:5525-5540. [PMID: 38206470 DOI: 10.1007/s12035-024-03920-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024]
Abstract
Immune response is pivotal in the secondary injury of spinal cord injury (SCI). Polarization of macrophages (MΦ) influences the immune response in the secondary injury, which is regulated by several immune-related proteins. M2Φ plays the immunomodulatory role in the central nervous system. This study used bioinformatic analysis and machine algorithms to screen hub immune-related proteins after SCI and experimentally investigate the role of the target protein in the M2Φ polarization and immunomodulation in rats and in vitro after SCI. We downloaded GSE151371 and GSE45006, hub immune-related genes were screened using machine learning algorithms, and the expression of S100A9 was verified by datasets. Allen's weight-drop injury SCI model in Sprague-Dawley rat and bone marrow-derived rat MΦ with myelin debris model were used to study the effects of S100A9 on M2Φ polarization and immunomodulation at the lesion site and in vitro. Bioinformatic analysis showed that S100A9 acts as a hub immune-related gene in the SCI patients and rats. S100A9 increased at the lesion site in SCI rats, and its inhibition reduced CD206 and ARG-1 expression. Exogenous S100A9 promoted CD206 and ARG-1 expression in MΦ. S100A9 also increased the expression of PD-L1 and decreased MHC II at the lesion site in SCI rats and MΦ with myelin debris, and enhanced mitochondrial activity in rat MΦ with myelin debris. In conclusion, S100A9 is an indispensable factor in the immune process in secondary injury following SCI.
Collapse
Affiliation(s)
- Junqiao Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zhiqiang Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Beiyang Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Chen Deng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wei Wang
- Department of Urology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Lin Sun
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
6
|
Liu X, Li Z, Tong J, Wu F, Jin H, Liu K. Characterization of the Expressions and m6A Methylation Modification Patterns of mRNAs and lncRNAs in a Spinal Cord Injury Rat Model. Mol Neurobiol 2024:10.1007/s12035-024-04297-z. [PMID: 38907070 DOI: 10.1007/s12035-024-04297-z] [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: 02/02/2024] [Accepted: 06/09/2024] [Indexed: 06/23/2024]
Abstract
Spinal cord injury (SCI) is a serious central nervous system disease with no effective treatment strategy presently due to its complex pathogenic mechanism. N6-methyladenosine (m6A) methylation modification plays an important role in diverse physiological and pathological processes. However, our understanding of the potential mechanisms of messenger RNA (mRNA) and long non-coding RNAs (lncRNA) m6A methylation in SCI is currently limited. Here, comprehensive m6A profiles and gene expression patterns of mRNAs and lncRNAs in spinal cord tissues after SCI were identified using microarray analysis of immunoprecipitated methylated RNAs. A total of 3745 mRNAs (2343 hypermethylated and 1402 hypomethylated) and 738 lncRNAs (488 hypermethylated and 250 hypomethylated) were differentially methylated with m6A modifications in the SCI and sham rats. Functional analysis revealed that differentially m6A-modified mRNAs were mainly involved in immune inflammatory response, nervous system development, and focal adhesion pathway. In contrast, differentially m6A-modified lncRNAs were mainly related to antigen processing and presentation, the apoptotic process, and the mitogen-activated protein kinases (MAPKs) signaling pathway. In addition, combined analysis of m6A methylation and RNA expression results revealed that 1636 hypermethylated mRNAs and 262 hypermethylated lncRNAs were up-regulated, and 1571 hypomethylated mRNAs and 204 lncRNAs were down-regulated. Furthermore, we validated the altered levels of m6A methylation and RNA expression of five mRNAs (CD68, Gpnmb, Lilrb4, Lamp5, and Snap25) and five lncRNAs (XR_360518, uc.393 + , NR_131064, uc.280 - , and XR_597251) using MeRIP-qPCR and qRT-PCR. This study expands our understanding of the molecular mechanisms underlying m6A modification in SCI and provides novel insights to promote functional recovery after SCI.
Collapse
Affiliation(s)
- Xin Liu
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, 518040, Guangdong, China
| | - Zhiling Li
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Juncheng Tong
- Shenzhen Luohu Hospital Group, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Fan Wu
- Shenzhen Luohu Hospital Group, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China
| | - Hui Jin
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, Guangdong, China.
- Research Centre, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Kaiqing Liu
- Shenzhen Luohu Hospital Group, The Third Affiliated Hospital of Shenzhen University, Shenzhen, 518000, China.
| |
Collapse
|
7
|
Miao X, Lin J, Li A, Gao T, Liu T, Shen J, Sun Y, Wei J, Bao B, Zheng X. AAV-mediated VEGFA overexpression promotes angiogenesis and recovery of locomotor function following spinal cord injury via PI3K/Akt signaling. Exp Neurol 2024; 375:114739. [PMID: 38401852 DOI: 10.1016/j.expneurol.2024.114739] [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: 11/16/2023] [Revised: 02/09/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Spinal cord injury (SCI) is a disorder of the central nervous system resulting from various factors such as trauma, inflammation, tumors, and other etiologies. This condition leads to impairment in motor, sensory, and autonomic functions below the level of injury. Limitations of current therapeutic approaches prompt an investigation into therapeutic angiogenesis through persistent local expression of proangiogenic factors. Here, we investigated whether overexpression of adeno-associated virus (AAV)-mediated vascular endothelial growth factor A (VEGFA) in mouse SCI promoted locomotor function recovery, and whether the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway was mechanistically involved. Three weeks before SCI, AAV-VEGFA was injected at the T10 level to induce VEGFA overexpression. Neurofunctional, histological, and biochemical assessments were done to determine tissue damage and/or recovery of neuromuscular and behavioral impairments. Daily injections of the PI3K/Akt pathway inhibitor LY294002 were made to assess a possible mechanism. AAV-VEGFA overexpression dramatically improved locomotor function and ameliorated pathological injury caused by SCI. Improved motor-evoked potentials in hindlimbs and more spinal CD31-positive microvessels were observed in AAV-VEGFA-overexpressing mice. LY294002 reduced PI3K and Akt phosphorylation levels and attenuated AAV-VEGFA-related improvements. In conclusion, sustained local AAV-mediated VEGFA overexpression in spinal cord can significantly promote angiogenesis and ameliorate locomotor impairment after SCI in a contusion mouse model through activation of the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xin Miao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Junqing Lin
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Ang Li
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Tao Gao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Tiexin Liu
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Junjie Shen
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Yi Sun
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Jiabao Wei
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Bingbo Bao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China
| | - Xianyou Zheng
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Center for Orthopaedics, Shanghai, China.
| |
Collapse
|
8
|
Gu C, Geng X, Wu Y, Dai Y, Zeng J, Wang Z, Fang H, Sun Y, Chen X. Engineered Macrophage Membrane-Coated Nanoparticles with Enhanced CCR2 Expression Promote Spinal Cord Injury Repair by Suppressing Neuroinflammation and Neuronal death. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305659. [PMID: 37884477 DOI: 10.1002/smll.202305659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Spinal cord injury (SCI) is a severe neurological disorder characterized by significant disability and limited treatment options. Mitigating the secondary inflammatory response following the initial injury is the primary focus of current research in the treatment of SCI. CCL2 (C─C motif chemokine ligand 2) serves as the primary regulator responsible for inflammatory chemotaxis of the majority of peripheral immune cells, blocking the CCL2-CCR2 (C─C chemokine receptor type 2) axis has shown considerable therapeutic potential for inflammatory diseases, including SCI. In this study, it presents a multifunctional biomimetic nanoplatform (CCR2-MM@PLGA/Cur) specifically designed to target the CCL2-CCR2 axis, which consisted of an engineered macrophage membrane (MM) coating with enhanced CCR2 expression and a PLGA (poly (lactic-co-glycolic acid)) nanoparticle that encapsulated therapeutic drugs. CCR2 overexpression on MM not only enhanced drug-targeted delivery to the injury site, but also attenuated macrophage infiltration, microglia pro-inflammatory polarization, and neuronal apoptosis by trapping CCL2. Consequently, it facilitated neural regeneration and motor function recovery in SCI mice, enabling a comprehensive treatment approach for SCI. The feasibility and efficacy of this platform are confirmed through a series of in vitro and in vivo assays, offering new insights and potential avenues for further exploration in the treatment of SCI.
Collapse
Affiliation(s)
- Changjiang Gu
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Xiangwu Geng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Yicheng Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Yuya Dai
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Junkai Zeng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Zhenqiang Wang
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Huapan Fang
- State Key Laboratory of Physical Chemistry of Solid Surfaces College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, P. R. China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen, 361005, P. R. China
| | - Yanqing Sun
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, P. R. China
| | - Xiongsheng Chen
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
- Department of Orthopaedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Shanghai, 200080, P. R. China
| |
Collapse
|
9
|
Fang S, Tang H, Li HL, Han TC, Li ZJ, Yin ZS, Chu JJ. CCL2 Knockdown Attenuates Inflammatory Response After Spinal Cord Injury Through the PI3K/Akt Signaling Pathway: Bioinformatics Analysis and Experimental Validation. Mol Neurobiol 2024; 61:1433-1447. [PMID: 37721689 DOI: 10.1007/s12035-023-03641-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Spinal cord injury (SCI) is a common clinical problem in orthopedics with a lack of effective treatments and drug targets. In the present study, we performed bioinformatic analysis of SCI datasets GSE464 and GSE45006 in the Gene Expression Omnibus (GEO) public database and experimentally validated CCL2 expression in an animal model of SCI. This was followed by stimulation of PC-12 cells using hydrogen peroxide to construct a cellular model of SCI. CCL2 expression was knocked down using small interfering RNA (si-CCL2), and PI3K signaling pathway inhibitors and activators were used to validate and observe the changes in downstream inflammation. Through data mining, we found that the inflammatory chemokine CCL2 and PI3K/Akt signaling pathways after SCI expression were significantly increased, and after peroxide stimulation of PC-12 cells with CCL2 knockdown, their downstream cellular inflammatory factor levels were decreased. The PI3K/Akt signaling pathway was blocked by PI3K inhibitors, and the downstream inflammatory response was suppressed. In contrast, when PI3K activators were used, the inflammatory response was enhanced, indicating that the CCL2-PI3K/Akt signaling pathway plays a key role in the regulation of the inflammatory response. This study revealed that the inflammatory chemokine CCL2 can regulate the inflammatory response of PC-12 cells through the PI3K/Akt signaling pathway, and blocking the expression of the inflammatory chemokine CCL2 may be a promising strategy for the treatment of secondary injury after SCI.
Collapse
Affiliation(s)
- Sheng Fang
- School of Medicine, Lishui University, Lishui, 323000, China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China
| | - Hai-Long Li
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Ti-Chao Han
- Department of Orthopedics, The Linquan County People's Hospital, 109 Tong Yang Road, Fuyang, Anhui Province, 236400, People's Republic of China
| | - Zi-Jie Li
- Department of Anesthesiology, The Linquan County People's Hospital, 109 Tong Yang Road, Fuyang, Anhui Province, 236400, People's Republic of China
| | - Zong-Sheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China.
| | - Jian-Jun Chu
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China.
| |
Collapse
|
10
|
Mukherjee S, Bruno MEC, Oakes J, Hawk GS, Stromberg AJ, Cohen DA, Starr ME. Mechanisms of γδ T cell accumulation in visceral adipose tissue with aging. FRONTIERS IN AGING 2024; 4:1258836. [PMID: 38274288 PMCID: PMC10808514 DOI: 10.3389/fragi.2023.1258836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024]
Abstract
γδ T cells are resident in visceral adipose tissue (VAT) where they show an age-associated increase in numbers and contribute to local and systemic chronic inflammation. However, regulation of this population and mechanisms for the age-dependent accumulation are not known. In this study, we identified a progressive trend of γδ T cell accumulation in VAT over the lifespan in mice and explored physiological mechanisms contributing to accumulation. Using isochronic parabiotic pairs of wild-type (WT) and T cell receptor delta knockout (TCRδ KO) mice at young and old age, we confirmed that VAT γδ T cells are predominately a tissue-resident population which is sustained in aging. Migration of peripheral γδ T cells into VAT was observed at less than 10%, with a decreasing trend by aging, suggesting a minor contribution of recruitment to γδ T cell accumulation with aging. Since tissue-resident T cell numbers are tightly regulated by a balance between proliferation and programmed cell death, we further explored these processes. Using in vivo EdU incorporation and the proliferation marker Ki67, we found that the absolute number of proliferating γδ T cells in VAT is significantly higher in the aged compared to young and middle-aged mice, despite a decline in the proportion of proliferating to non-proliferating cells by age. Analysis of apoptosis via caspase 3/7 activation revealed that VAT γδ T cells show reduced apoptosis starting at middle age and continuing into old age. Further, induction of apoptosis using pharmacological inhibitors of Bcl2 family proteins revealed that VAT γδ T cells at middle age are uniquely protected from apoptosis via a mechanism independent of traditional anti-apoptotic Bcl2-family proteins. Collectively, these data indicate that protection from apoptosis at middle age increases survival of tissue-resident γδ T cells resulting in an increased number of proliferative cells from middle age onward, and leading to the age-associated accumulation of γδ T cells in VAT. These findings are important to better understand how adipose tissue dysfunction and related changes in the immune profile contribute to inflammaging among the elderly.
Collapse
Affiliation(s)
- Sujata Mukherjee
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Maria E. C. Bruno
- Division of Research, Department of Surgery, University of Kentucky, Lexington, KY, United States
| | - Jason Oakes
- Division of Laboratory Animal Resources, University of Kentucky, Lexington, KY, United States
| | - Gregory S. Hawk
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Arnold J. Stromberg
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY, United States
| | - Donald A. Cohen
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Marlene E. Starr
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
- Division of Research, Department of Surgery, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
11
|
Xi D, Liu P, Feng Y, Teng Y, Liang Y, Zhou J, Deng H, Zeng G, Zong S. Fecal microbiota transplantation regulates the microbiota-gut-spinal cord axis to promote recovery after spinal cord injury. Int Immunopharmacol 2024; 126:111212. [PMID: 37979452 DOI: 10.1016/j.intimp.2023.111212] [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: 09/20/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
Spinal cord injury (SCI) is devastating for patients, and currently lacks effective treatments. Dysbiosis commonly occurs after SCI and has significant immunomodulatory effects, but its impact on recovery remains unclear. The current study investigated the effects and mechanisms of fecal microbiota transplantation (FMT) in SCI. FMT was administered in a rat model of SCI and spinal pathology, inflammatory cytokines, and gut microbiome composition were assessed. Flow cytometry identified a source of interleukin (IL)-17 in spinal cord tissues, and carboxyfluorescein succimidyl ester labeling tracked γδ T cell migration. In vitro coculture was used to analyze the regulatory mechanisms of γδ T cells. Seahorse analysis was used to profile dendritic cell (DC) metabolism. Here we show that FMT improved spinal pathology and dampened post-injury inflammation. It also corrected post-SCI dysbiosis, increasing levels of the beneficial bacterium Akkermansia. The therapeutic effects of FMT were mediated by IL-17 produced by γδ T cells. FMT regulated γδ T cells via DC-T regulatory cell interaction, and induced metabolic reprogramming in DCs. These findings suggest that FMT represents a promising therapeutic approach for SCI, with potential to target IL-17+ γδ T cells. Elucidating the interconnected pathways between microbiota, immunity, and the spinal cord may facilitate novel treatment strategies.
Collapse
Affiliation(s)
- Deshuang Xi
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Pan Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, He-nan, China
| | - Yanbing Feng
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yilin Teng
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yu Liang
- Department of Spine Surgery, The Second People's Hospital of Nanning, Nanning 530021, Guangxi, China
| | - Junhong Zhou
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Hao Deng
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Gaofeng Zeng
- College of Public Hygiene of Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Shaohui Zong
- Department of Spine and Osteopathy Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
12
|
Li Y, Chen Y, Hu X, Ouyang F, Li J, Huang J, Ye J, Shan F, Luo Y, Yu S, Li Z, Yao F, Liu Y, Shi Y, Zheng M, Cheng L, Jing J. Fingolimod (FTY720) Hinders Interferon-γ-Mediated Fibrotic Scar Formation and Facilitates Neurological Recovery After Spinal Cord Injury. J Neurotrauma 2023; 40:2580-2595. [PMID: 36879472 DOI: 10.1089/neu.2022.0387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Following spinal cord injury (SCI), fibrotic scar inhibits axon regeneration and impairs neurological function recovery. It has been reported that T cell-derived interferon (IFN)-γ plays a pivotal role in promoting fibrotic scarring in neurodegenerative disease. However, the role of IFN-γ in fibrotic scar formation after SCI has not been declared. In this study, a spinal cord crush injury mouse was established. Western blot and immunofluorescence showed that IFN-γ was surrounded by fibroblasts at 3, 7, 14, and 28 days post-injury. Moreover, IFN-γ is mainly secreted by T cells after SCI. Further, in situ injection of IFN-γ into the normal spinal cord resulted in fibrotic scar formation and inflammation response at 7 days post-injection. After SCI, the intraperitoneal injection of fingolimod (FTY720), a sphingosine-1-phosphate receptor 1 (S1PR1) modulator and W146, an S1PR1 antagonist, significantly reduced T cell infiltration, attenuating fibrotic scarring via inhibiting IFN-γ/IFN-γR pathway, while in situ injection of IFN-γ diminished the effect of FTY720 on reducing fibrotic scarring. FTY720 treatment inhibited inflammation, decreased lesion size, and promoted neuroprotection and neurological recovery after SCI. These findings demonstrate that the inhibition of T cell-derived IFN-γ by FTY720 suppressed fibrotic scarring and contributed to neurological recovery after SCI.
Collapse
Affiliation(s)
- Yiteng Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihao Chen
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xuyang Hu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangru Ouyang
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jianjian Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jinxin Huang
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jianan Ye
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fangli Shan
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yong Luo
- Scientific Research and Experiment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuisheng Yu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ziyu Li
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fei Yao
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanchang Liu
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi Shi
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meige Zheng
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Cheng
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juehua Jing
- Department of Orthopedics, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Institute of Orthopedics, Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
13
|
Estera LA, Walsh SP, Headen JA, Williamson RE, Kalinski AL. Neuroinflammation: Breaking barriers and bridging gaps. Neurosci Res 2023; 197:9-17. [PMID: 34748905 DOI: 10.1016/j.neures.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/04/2023]
Abstract
Neurons are the cells of the nervous system and are responsible for every thought, movement and perception. Immune cells are the cells of the immune system, constantly protecting from foreign pathogens. Understanding the interaction between the two systems is especially important in disease states such as autoimmune or neurodegenerative disease. Unfortunately, this interaction is typically detrimental to the host. However, recent efforts have focused on how neurons and immune cells interact, either directly or indirectly, following traumatic injury to the nervous system. The outcome of this interaction can be beneficial - leading to successful neural repair, or detrimental - leading to functional deficits, depending on where the injury occurs. This review will discuss our understanding of neuron-immune cell interactions after traumatic injury to both the peripheral and central nervous system.
Collapse
Affiliation(s)
- Lora A Estera
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Sam P Walsh
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Jordan A Headen
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | | | - Ashley L Kalinski
- Department of Biology, Ball State University, Muncie, IN 47306, USA.
| |
Collapse
|
14
|
Conedera FM, Runnels JM, Stein JV, Alt C, Enzmann V, Lin CP. Assessing the role of T cells in response to retinal injury to uncover new therapeutic targets for the treatment of retinal degeneration. J Neuroinflammation 2023; 20:206. [PMID: 37689689 PMCID: PMC10492418 DOI: 10.1186/s12974-023-02867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/31/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Retinal degeneration is a disease affecting the eye, which is an immune-privileged site because of its anatomical and physiological properties. Alterations in retinal homeostasis-because of injury, disease, or aging-initiate inflammatory cascades, where peripheral leukocytes (PL) infiltrate the parenchyma, leading to retinal degeneration. So far, research on PL's role in retinal degeneration was limited to observing a few cell types at specific times or sectioning the tissue. This restricted our understanding of immune cell interactions and response duration. METHODS In vivo microscopy in preclinical mouse models can overcome these limitations enabling the spatio-temporal characterization of PL dynamics. Through in vivo imaging, we assessed structural and fluorescence changes in response to a focal injury at a defined location over time. We also utilized minimally invasive techniques, pharmacological interventions, and knockout (KO) mice to determine the role of PL in local inflammation. Furthermore, we investigated PL abundance and localization during retinal degeneration in human eyes by histological analysis to assess to which extent our preclinical study translates to human retinal degeneration. RESULTS We demonstrate that PL, especially T cells, play a detrimental role during retinal injury response. In mice, we observed the recruitment of helper and cytotoxic T cells in the parenchyma post-injury, and T cells also resided in the macula and peripheral retina in pathological conditions in humans. Additionally, we found that the pharmacological PL reduction and genetic depletion of T-cells reduced injured areas in murine retinas and rescued the blood-retina barrier (BRB) integrity. Both conditions promoted morphological changes of Cx3cr1+ cells, including microglial cells, toward an amoeboid phenotype during injury response. Interestingly, selective depletion of CD8+ T cells accelerated recovery of the BRB compared to broader depletions. After anti-CD8 treatment, the retinal function improved, concomitant to a beneficial immune response. CONCLUSIONS Our data provide novel insights into the adaptive immune response to retinal injury in mice and human retinal degeneration. Such information is fundamental to understanding retinal disorders and developing therapeutics to modulate immune responses to retinal degeneration safely.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
- Department of Ophthalmology, Bern University Hospital, Bern, Switzerland
| | - Judith M Runnels
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital, Bern, Switzerland.
- Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Zhang B, Hu M, Ma Q, Li K, Li X, He X, Shu P, Chen Y, Gao G, Qin D, Guo F, Zhao J, Liu N, Zhou K, Feng M, Liao W, Li D, Wang X, Wang Y. Optimized CAR-T therapy based on spatiotemporal changes and chemotactic mechanisms of MDSCs induced by hypofractionated radiotherapy. Mol Ther 2023; 31:2105-2119. [PMID: 37073129 PMCID: PMC10362417 DOI: 10.1016/j.ymthe.2023.04.008] [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: 09/20/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 04/20/2023] Open
Abstract
Poor intratumoral infiltration is the major challenge for chimeric antigen receptor (CAR)-T cell therapy in solid tumors. Hypofractionated radiotherapy (HFRT) has been reported to induce immune cell infiltration and reshape the tumor immune microenvironment. Here, we showed that HFRT (5 × 5 Gy) mediated an early accumulation of intratumoral myeloid-derived suppressor cells (MDSCs) and decreased infiltration of T cells in the tumor microenvironment (TME) of immunocompetent mice bearing triple-negative breast cancer (TNBC) or colon cancer, which was further confirmed in tumors from patients. RNA sequencing (RNA-seq) and cytokine profiling analysis revealed that HFRT induced the activation and proliferation of tumor-infiltrated MDSCs, which was mediated by the interactions of multiple chemokines and chemokine receptors. Further investigation showed that when combined with HFRT, CXCR2 blockade significantly inhibited MDSCs trafficking to tumors and effectively enhanced the intratumoral infiltration and treatment efficacy of CAR-T cells. Our study demonstrates that MDSCs blockade combined with HFRT is promising for CAR-T cell therapy optimization in solid tumors.
Collapse
Affiliation(s)
- Benxia Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Hu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Qizhi Ma
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kai Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xue Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xia He
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pei Shu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Chen
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Diyuan Qin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fuchun Guo
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jian Zhao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ning Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kexun Zhou
- Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - MingYang Feng
- Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weiting Liao
- Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, and Precision Medicine Research Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
16
|
Hu X, Xu W, Ren Y, Wang Z, He X, Huang R, Ma B, Zhao J, Zhu R, Cheng L. Spinal cord injury: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:245. [PMID: 37357239 DOI: 10.1038/s41392-023-01477-6] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 06/27/2023] Open
Abstract
Spinal cord injury (SCI) remains a severe condition with an extremely high disability rate. The challenges of SCI repair include its complex pathological mechanisms and the difficulties of neural regeneration in the central nervous system. In the past few decades, researchers have attempted to completely elucidate the pathological mechanism of SCI and identify effective strategies to promote axon regeneration and neural circuit remodeling, but the results have not been ideal. Recently, new pathological mechanisms of SCI, especially the interactions between immune and neural cell responses, have been revealed by single-cell sequencing and spatial transcriptome analysis. With the development of bioactive materials and stem cells, more attention has been focused on forming intermediate neural networks to promote neural regeneration and neural circuit reconstruction than on promoting axonal regeneration in the corticospinal tract. Furthermore, technologies to control physical parameters such as electricity, magnetism and ultrasound have been constantly innovated and applied in neural cell fate regulation. Among these advanced novel strategies and technologies, stem cell therapy, biomaterial transplantation, and electromagnetic stimulation have entered into the stage of clinical trials, and some of them have already been applied in clinical treatment. In this review, we outline the overall epidemiology and pathophysiology of SCI, expound on the latest research progress related to neural regeneration and circuit reconstruction in detail, and propose future directions for SCI repair and clinical applications.
Collapse
Affiliation(s)
- Xiao Hu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Wei Xu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Yilong Ren
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Zhaojie Wang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Xiaolie He
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Runzhi Huang
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Bei Ma
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Jingwei Zhao
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China
| | - Rongrong Zhu
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, 200065, Shanghai, China.
- Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, 200065, Shanghai, China.
- Clinical Center For Brain And Spinal Cord Research, Tongji University, 200065, Shanghai, China.
| |
Collapse
|
17
|
Maheshwari S, Dwyer LJ, Sîrbulescu RF. Inflammation and immunomodulation in central nervous system injury - B cells as a novel therapeutic opportunity. Neurobiol Dis 2023; 180:106077. [PMID: 36914074 PMCID: PMC10758988 DOI: 10.1016/j.nbd.2023.106077] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
Acute injury to the central nervous system (CNS) remains a complex and challenging clinical need. CNS injury initiates a dynamic neuroinflammatory response, mediated by both resident and infiltrating immune cells. Following the primary injury, dysregulated inflammatory cascades have been implicated in sustaining a pro-inflammatory microenvironment, driving secondary neurodegeneration and the development of lasting neurological dysfunction. Due to the multifaceted nature of CNS injury, clinically effective therapies for conditions such as traumatic brain injury (TBI), spinal cord injury (SCI), and stroke have proven challenging to develop. No therapeutics that adequately address the chronic inflammatory component of secondary CNS injury are currently available. Recently, B lymphocytes have gained increasing appreciation for their role in maintaining immune homeostasis and regulating inflammatory responses in the context of tissue injury. Here we review the neuroinflammatory response to CNS injury with particular focus on the underexplored role of B cells and summarize recent results on the use of purified B lymphocytes as a novel immunomodulatory therapeutic for tissue injury, particularly in the CNS.
Collapse
Affiliation(s)
- Saumya Maheshwari
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liam J Dwyer
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Liu P, Liu M, Xi D, Bai Y, Ma R, Mo Y, Zeng G, Zong S. Short-chain fatty acids ameliorate spinal cord injury recovery by regulating the balance of regulatory T cells and effector IL-17 + γδ T cells. J Zhejiang Univ Sci B 2023; 24:312-325. [PMID: 37056207 PMCID: PMC10106403 DOI: 10.1631/jzus.b2200417] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/30/2022] [Indexed: 04/15/2023]
Abstract
Spinal cord injury (SCI) causes motor, sensory, and autonomic dysfunctions. The gut microbiome has an important role in SCI, while short-chain fatty acids (SCFAs) are one of the main bioactive mediators of microbiota. In the present study, we explored the effects of oral administration of exogenous SCFAs on the recovery of locomotor function and tissue repair in SCI. Allen's method was utilized to establish an SCI model in Sprague-Dawley (SD) rats. The animals received water containing a mixture of 150 mmol/L SCFAs after SCI. After 21 d of treatment, the Basso, Beattie, and Bresnahan (BBB) score increased, the regularity index improved, and the base of support (BOS) value declined. Spinal cord tissue inflammatory infiltration was alleviated, the spinal cord necrosis cavity was reduced, and the numbers of motor neurons and Nissl bodies were elevated. Enzyme-linked immunosorbent assay (ELISA), real-time quantitative polymerase chain reaction (qPCR), and immunohistochemistry assay revealed that the expression of interleukin (IL)-10 increased and that of IL-17 decreased in the spinal cord. SCFAs promoted gut homeostasis, induced intestinal T cells to shift toward an anti-inflammatory phenotype, and promoted regulatory T (Treg) cells to secrete IL-10, affecting Treg cells and IL-17+ γδ T cells in the spinal cord. Furthermore, we observed that Treg cells migrated from the gut to the spinal cord region after SCI. The above findings confirm that SCFAs can regulate Treg cells in the gut and affect the balance of Treg and IL-17+ γδ T cells in the spinal cord, which inhibits the inflammatory response and promotes the motor function in SCI rats. Our findings suggest that there is a relationship among gut, spinal cord, and immune cells, and the "gut-spinal cord-immune" axis may be one of the mechanisms regulating neural repair after SCI.
Collapse
Affiliation(s)
- Pan Liu
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics, the Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, China
| | - Mingfu Liu
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Deshuang Xi
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yiguang Bai
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics, Nanchong Central Hosipital, the Second Clinical Institute of North Sichuan Medical College, Nanchong 637000, China
| | - Ruixin Ma
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, China
| | - Yaomin Mo
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Gaofeng Zeng
- College of Public Hygiene of Guangxi Medical University, Nanning 530021, China.
| | - Shaohui Zong
- Department of Spine Osteopathic, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
19
|
Fang S, Tang H, Li MZ, Chu JJ, Yin ZS, Jia QY. Identification of the CCL2 PI3K/Akt axis involved in autophagy and apoptosis after spinal cord injury. Metab Brain Dis 2023; 38:1335-1349. [PMID: 36795287 DOI: 10.1007/s11011-023-01181-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/04/2023] [Indexed: 02/17/2023]
Abstract
Spinal cord injury (SCI) is a devastating neurological disease with no cure that usually results in irreversible loss of sensory and voluntary motor functions below the injury site. We conducted an in-depth bioinformatics analysis combining the gene expression omnibus spinal cord injury database and the autophagy database and found that the expression of the autophagy gene CCL2 was significantly upregulated and the PI3K/Akt/mTOR signaling pathway was activated after SCI. The results of the bioinformatics analysis were verified by constructing animal and cellular models of SCI. We then used small interfering RNA to inhibit the expression of CCL2 and PI3K to inhibit and activate the PI3K/Akt/mTOR signaling pathway; western blot, immunofluorescence, monodansylcadaverine, and cell flow techniques were used to detect the expression of key proteins involved in downstream autophagy and apoptosis. We found that when PI3K inhibitors were activated, apoptosis decreased, the levels of autophagy-positive proteins LC3-I/LC3-II and Bcl-1 increased, the levels of autophagy-negative protein P62 decreased, the levels of pro-apoptotic proteins Bax and caspase-3 decreased, the levels of the apoptosis-inhibiting protein Bcl-2 increased. In contrast, when a PI3K activator was used, autophagy was inhibited, and apoptosis was increased. This study revealed the effect of CCL2 on autophagy and apoptosis after SCI through the PI3K/Akt/mTOR signaling pathway. By blocking the expression of the autophagy-related gene CCL2, the autophagic protective response can be activated, and apoptosis can be inhibited, which may be a promising strategy for the treatment of SCI.
Collapse
Affiliation(s)
- Sheng Fang
- Department of Orthopedics, Hefei hospital Affiliated to Anhui Medical University, Guang De Road, 230011, Hefei, Anhui Province, People's Republic of China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, 230022, Hefei, China
| | - Ming-Zhi Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, 230022, Hefei, China
- Department of Orthopedics, the Linquan county people's Hospital Affiliated to Anhui Medical University, 109 Tongyang Road, 236400, Fuyang, Anhui Province, People's Republic of China
| | - Jian-Jun Chu
- Department of Orthopedics, Hefei hospital Affiliated to Anhui Medical University, Guang De Road, 230011, Hefei, Anhui Province, People's Republic of China
| | - Zong-Sheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, 230022, Hefei, China.
| | - Qi-Yu Jia
- Department of Orthopedics, Hefei hospital Affiliated to Anhui Medical University, Guang De Road, 230011, Hefei, Anhui Province, People's Republic of China.
| |
Collapse
|
20
|
[Water tank scale: a reliable method for assessing motor function after spinal cord injury in rats]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:99-104. [PMID: 36856216 DOI: 10.12122/j.issn.1673-4254.2023.01.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
OBJECTIVE To analyze the reliability of the Water Tank Scale for assessing recovery of motor function after spinal cord injury (SCI) in rats. METHODS Thirty-six adult female SD rats were randomly divided into SCI and sham-operated groups (n= 18). The recovery of the hind limb motor function was assessed using Water Tank scoring, BBB scoring, and motor-evoked potentials (MEP) at 1, 3, 5, 7, 14 and 21 days after SCI. MEP was used as the gold standard for analyzing and comparing differences between the two scoring methods. RESULTS The Water Tank scores of the rats were significantly higher than the BBB scores on day 3 (0.22±0.43 vs 0, P < 0.05) and also on days 5, 7 and 14 after SCI (0.67±0.49 vs 0.11±0.32, 4.33±1.19 vs 2.83±1.04, 8.61± 1.20 vs 7.06±1.0, P < 0.01). On day 21 after SCI, the scores of the Water Tank Scale of the rats did not significantly differ from the BBB scores (14.78±1.06 vs 14.50±1.47, P>0.05). Neurophysiological monitoring showed that both the Water Tank score and BBB score were significantly correlated with MEP latency, but the Water Tank score had a greater correlation coefficient with MEP latency (r=-0.90). CONCLUSION Compared with the BBB scale, Water Tank scoring allows more objective and accurate assessment of functional recovery of the spinal cord in early stages following SCI in rats, and can thus be used as a reliable method for assessing functional recovery of the hind limbs in rat models of acute SCI.
Collapse
|
21
|
Tang H, Gu Y, Jiang L, Zheng G, Pan Z, Jiang X. The role of immune cells and associated immunological factors in the immune response to spinal cord injury. Front Immunol 2023; 13:1070540. [PMID: 36685599 PMCID: PMC9849245 DOI: 10.3389/fimmu.2022.1070540] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological condition prevalent worldwide. Where the pathological mechanisms underlying SCI are concerned, we can distinguish between primary injury caused by initial mechanical damage and secondary injury characterized by a series of biological responses, such as vascular dysfunction, oxidative stress, neurotransmitter toxicity, lipid peroxidation, and immune-inflammatory response. Secondary injury causes further tissue loss and dysfunction, and the immune response appears to be the key molecular mechanism affecting injured tissue regeneration and functional recovery from SCI. Immune response after SCI involves the activation of different immune cells and the production of immunity-associated chemicals. With the development of new biological technologies, such as transcriptomics, the heterogeneity of immune cells and chemicals can be classified with greater precision. In this review, we focus on the current understanding of the heterogeneity of these immune components and the roles they play in SCI, including reactive astrogliosis and glial scar formation, neutrophil migration, macrophage transformation, resident microglia activation and proliferation, and the humoral immunity mediated by T and B cells. We also summarize findings from clinical trials of immunomodulatory therapies for SCI and briefly review promising therapeutic drugs currently being researched.
Collapse
Affiliation(s)
- Huaguo Tang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yuanjie Gu
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Lei Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Gang Zheng
- Department of Neurosurgery, The Central Hospital Affiliated to Shaoxing University, Jiaxing, China
| | - Zhuoer Pan
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xiugui Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
22
|
Transplantation of olfactory ensheathing cells decreases local and serological monocyte chemoattractant protein 1 level during the acute phase of rat spinal cord injury. Neuroreport 2022; 33:729-741. [PMID: 36250430 DOI: 10.1097/wnr.0000000000001839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Monocyte chemoattractant protein 1 (MCP1) is one of the most upregulated cytokines in the spinal cord and serum throughout acute spinal cord injury (SCI). Olfactory ensheathing cells (OECs) transplantation improves SCI through multiple mechanisms, including immunomodulation. Our study aimed to investigate whether OECs ameliorate acute inflammation after SCI by modulating MCP1 expression. METHODS We established a standardized clinically relevant contusion model using the NYU impactor. OECs were administered to the injured spinal cord via microinjection 30 minutes after injury. Rat locomotor functions were assessed by the Basso-Beattie-Bresnahan scale score. Time-course histopathological (H&E and IHC) analyses were performed to record rapid changes in acute inflammation at lesion epicenters. Serum MCP1 level was detected by ELISA assay. RESULTS BBB scores showed improved locomotor functional recoveries in the OECs transplantation group after SCI ( P < 0.05). Staining of H&E and CD68 illustrated that OECs transplantation attenuated inflammatory response by reducing lesion areas and infiltrating myeloid cell numbers. We further revealed significantly decreased MCP1 levels in the spinal cord and serum after OECs transplantation ( P < 0.05). Noteworthily, distinct expression levels of MCP1 were found in rats undergoing a mild injury (cord impacted from a 10-mm height) compared to the moderate injury (25-mm) group. CONCLUSION Our study reports that transplantation of OECs promotes locomotor functional recovery after SCI and alleviates acute inflammation by decreasing local and serological MCP1 levels. We provide preliminary evidence that MCP1 might serve as a potential biomarker to reflect the severity of SCI, which is of great interest in future studies.
Collapse
|
23
|
Li J, Liu X, Wu H, Guo P, Li B, Wang J, Tian W, Chen D, Gao M, Zhou Z, Liu S. Identification of hub genes related to the innate immune response activated during spinal cord injury. FEBS Open Bio 2022; 12:1839-1856. [PMID: 36047918 PMCID: PMC9527585 DOI: 10.1002/2211-5463.13472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 08/01/2022] [Accepted: 08/17/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) often leads to sensory and motor dysfunction. Two major factors that hinder spinal cord repair are local inflammation and glial scar formation after SCI, and thus appropriate immunotherapy may alleviate damage. To characterize changes in gene expression that occur during SCI and thereby identify putative targets for immunotherapy, here we analyzed the dataset GSE5296 (containing one control group and six SCI groups at different timepoints) to identify differentially-expressed genes. Functional enrichment analysis was performed and a protein-protein interaction network was created to identify possible hub genes. Finally, we performed quantitative PCR to verify changes in gene expression. The CIBERSORT algorithm was used to analyze innate immune cell infiltration patterns. The dataset GSE162610 (containing one control group and three SCI groups at different timepoints) was analyzed to evaluate innate immune cell infiltration at the single-cell level. The dataset GSE151371 (containing one control group [n = 10] and an SCI group [n = 38]) was used to detect the expression of hub genes in the blood from SCI patients. Differentially-expressed innate immune-related genes at each timepoint were identified, and the functions and related signaling pathways of these genes were examined. Six hub genes were identified and verified. We then analyzed the expression characteristics of these hub genes and characteristics of innate immune infiltration in SCI; finally, we examined ligand expression in the context of the CCL signaling pathway and COMPLEMENT signaling pathway networks. This study reveals the characteristics of innate immune cell infiltration and temporal expression patterns of hub genes, and may aid in the development of immunotherapies for SCI.
Collapse
Affiliation(s)
- Jianfeng Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Huachuan Wu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Baoliang Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Jianmin Wang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Wei Tian
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopedics and TraumatologyBeijing Jishuitan HospitalChina
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopedics and TraumatologyBeijing Jishuitan HospitalChina
| | - Manman Gao
- Department of Sport Medicine, Institute of Translational MedicineThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalChina,Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical EngineeringShenzhen University Health Science CenterChina
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Shaoyu Liu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopedic Surgery, The Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopedic Research Institute/Department of Spinal SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
24
|
Wang C, Ma H, Zhang B, Hua T, Wang H, Wang L, Han L, Li Q, Wu W, Sun Y, Yang H, Lu X. Inhibition of IL1R1 or CASP4 attenuates spinal cord injury through ameliorating NLRP3 inflammasome-induced pyroptosis. Front Immunol 2022; 13:963582. [PMID: 35990672 PMCID: PMC9389052 DOI: 10.3389/fimmu.2022.963582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating trauma characterized by serious neuroinflammation and permanent neurological dysfunction. However, the molecular mechanism of SCI remains unclear, and few effective medical therapies are available at present. In this study, multiple bioinformatics methods were used to screen out novel targets for SCI, and the mechanism of these candidates during the progression of neuroinflammation as well as the therapeutic effects were both verified in a rat model of traumatic SCI. As a result, CASP4, IGSF6 and IL1R1 were identified as the potential diagnostic and therapeutic targets for SCI by computational analysis, which were enriched in NF-κB and IL6-JAK-STATA3 signaling pathways. In the injured spinal cord, these three signatures were up-regulated and closely correlated with NLRP3 inflammasome formation and gasdermin D (GSDMD) -induced pyroptosis. Intrathecal injection of inhibitors of IL1R1 or CASP4 improved the functional recovery of SCI rats and decreased the expression of these targets and inflammasome component proteins, such as NLRP3 and GSDMD. This treatment also inhibited the pp65 activation into the nucleus and apoptosis progression. In conclusion, our findings of the three targets shed new light on the pathogenesis of SCI, and the use of immunosuppressive agents targeting these proteins exerted anti-inflammatory effects against spinal cord inflammation by inhibiting NF-kB and NLRP3 inflammasome activation, thus blocking GSDMD -induced pyroptosis and immune activation.
Collapse
Affiliation(s)
- Chenfeng Wang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Hongdao Ma
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Bangke Zhang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Tong Hua
- Department of Anesthesiology, Shanghai Changzheng Hospital, Shanghai, China
| | - Haibin Wang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Liang Wang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Lin Han
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Qisheng Li
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Weiqing Wu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Yulin Sun
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
| | - Haisong Yang
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
- *Correspondence: Xuhua Lu, ; Haisong Yang,
| | - Xuhua Lu
- Department of Orthopaedics, Shanghai Changzheng Hospital, Shanghai, China
- *Correspondence: Xuhua Lu, ; Haisong Yang,
| |
Collapse
|
25
|
Li Y, Lei Z, Ritzel RM, He J, Li H, Choi HMC, Lipinski MM, Wu J. Impairment of autophagy after spinal cord injury potentiates neuroinflammation and motor function deficit in mice. Theranostics 2022; 12:5364-5388. [PMID: 35910787 PMCID: PMC9330534 DOI: 10.7150/thno.72713] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 01/25/2023] Open
Abstract
Autophagy is a catabolic process that degrades cytoplasmic constituents and organelles in the lysosome, thus serving an important role in cellular homeostasis and protection against insults. We previously reported that defects in autophagy contribute to neuronal cell damage in traumatic spinal cord injury (SCI). Recent data from other inflammatory models implicate autophagy in regulation of immune and inflammatory responses, with low levels of autophagic flux associated with pro-inflammatory phenotypes. In the present study, we examined the effects of genetically or pharmacologically manipulating autophagy on posttraumatic neuroinflammation and motor function after SCI in mice. Methods: Young adult male C57BL/6, CX3CR1-GFP, autophagy hypomorph Becn1+/- mice, and their wildtype (WT) littermates were subjected to moderate thoracic spinal cord contusion. Neuroinflammation and autophagic flux in the injured spinal cord were assessed using flow cytometry, immunohistochemistry, and NanoString gene expression analysis. Motor function was evaluated with the Basso Mouse Scale and horizontal ladder test. Lesion volume and spared white matter were evaluated by unbiased stereology. To stimulate autophagy, disaccharide trehalose, or sucrose control, was administered in the drinking water immediately after injury and for up to 6 weeks after SCI. Results: Flow cytometry demonstrated dysregulation of autophagic function in both microglia and infiltrating myeloid cells from the injured spinal cord at 3 days post-injury. Transgenic CX3CR1-GFP mice revealed increased autophagosome formation and inhibition of autophagic flux specifically in activated microglia/macrophages. NanoString analysis using the neuroinflammation panel demonstrated increased expression of proinflammatory genes and decreased expression of genes related to neuroprotection in Becn1+/- mice as compared to WT controls at 3 days post-SCI. These findings were further validated by qPCR, wherein we observed significantly higher expression of proinflammatory cytokines. Western blot analysis confirmed higher protein expression of the microglia/macrophage marker IBA-1, inflammasome marker, NLRP3, and innate immune response markers cGAS and STING in Becn1+/- mice at 3 day after SCI. Flow cytometry demonstrated that autophagy deficit did not affect either microglial or myeloid counts at 3 days post-injury, instead resulting in increased microglial production of proinflammatory cytokines. Finally, locomotor function showed significantly worse impairments in Becn1+/- mice up to 6 weeks after SCI, which was accompanied by worsening tissue damage. Conversely, treatment with a naturally occurring autophagy inducer trehalose, reduced protein levels of p62, an adaptor protein targeting cargo to autophagosomes as well as the NLRP3, STING, and IBA-1 at 3 days post-injury. Six weeks of trehalose treatment after SCI led to improved motor function recovery as compared to control group, which was accompanied by reduced tissue damage. Conclusions: Our data indicate that inhibition of autophagy after SCI potentiates pro-inflammatory activation in microglia and is associated with worse functional outcomes. Conversely, increasing autophagy with trehalose, decreased inflammation and improved outcomes. These findings highlight the importance of autophagy in spinal cord microglia and its role in secondary injury after SCI.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Hui Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Harry M C Choi
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Marta M Lipinski
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| |
Collapse
|
26
|
Cao L, Li Q. Revealing Potential Spinal Cord Injury Biomarkers and Immune Cell Infiltration Characteristics in Mice. Front Genet 2022; 13:883810. [PMID: 35706450 PMCID: PMC9189360 DOI: 10.3389/fgene.2022.883810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/29/2022] [Indexed: 08/04/2023] Open
Abstract
Spinal cord injury (SCI) is a disabling condition with significant morbidity and mortality. Currently, no effective SCI treatment exists. This study aimed to identify potential biomarkers and characterize the properties of immune cell infiltration during this pathological event. To eliminate batch effects, we concurrently analyzed two mouse SCI datasets (GSE5296, GSE47681) from the GEO database. First, we identified differentially expressed genes (DEGs) using linear models for microarray data (LIMMA) and performed functional enrichment studies on those DEGs. Next, we employed bioinformatics and machine-learning methods to identify and define the characteristic genes of SCI. Finally, we validated them using immunofluorescence and qRT-PCR. Additionally, this study assessed the inflammatory status of SCI by identifying cell types using CIBERSORT. Furthermore, we investigated the link between key markers and infiltrating immune cells. In total, we identified 561 robust DEGs. We identified Rab20 and Klf6 as SCI-specific biomarkers and demonstrated their significance using qRT-PCR in the mouse model. According to the examination of immune cell infiltration, M0, M1, and M2 macrophages, along with naive CD8, dendritic cell-activated, and CD4 Follicular T cells may have a role in the progression of SCI. Therefore, Rab20 and Klf6 could be accessible targets for diagnosing and treating SCI. Moreover, as previously stated, immune cell infiltration may significantly impact the development and progression of SCI.
Collapse
Affiliation(s)
- Liang Cao
- Department of Traumatic Orthopedics, The Second Affiliated Hospital, University of South China, Hengyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Qing Li
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
27
|
Sharma A, Noon JB, Kontodimas K, Garo LP, Platten J, Quinton LJ, Urban JF, Reinhardt C, Bosmann M. IL-27 Enhances γδ T Cell–Mediated Innate Resistance to Primary Hookworm Infection in the Lungs. THE JOURNAL OF IMMUNOLOGY 2022; 208:2008-2018. [PMID: 35354611 PMCID: PMC9012701 DOI: 10.4049/jimmunol.2000945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/09/2022] [Indexed: 11/19/2022]
Abstract
IL-27 is a heterodimeric IL-12 family cytokine formed by noncovalent association of the promiscuous EBI3 subunit and selective p28 subunit. IL-27 is produced by mononuclear phagocytes and unfolds pleiotropic immune-modulatory functions through ligation to IL-27 receptor α (IL-27RA). Although IL-27 is known to contribute to immunity and to limit inflammation after various infections, its relevance for host defense against multicellular parasites is still poorly defined. Here, we investigated the role of IL-27 during infection with the soil-transmitted hookworm, Nippostrongylus brasiliensis, in its early host intrapulmonary life cycle. IL-27(p28) was detectable in bronchoalveolar lavage fluid of C57BL/6J wild-type mice on day 1 after s.c. inoculation. IL-27RA expression was most abundant on lung-invading γδ T cells. Il27ra-/- mice showed increased lung parasite burden together with aggravated pulmonary hemorrhage and higher alveolar total protein leakage as a surrogate for epithelial-vascular barrier disruption. Conversely, injections of recombinant mouse (rm)IL-27 into wild-type mice reduced lung injury and parasite burden. In multiplex screens, higher airway accumulations of IL-6, TNF-α, and MCP-3 (CCL7) were observed in Il27ra-/- mice, whereas rmIL-27 treatment showed a reciprocal effect. Importantly, γδ T cell numbers in airways were enhanced by endogenous or administered IL-27. Further analysis revealed a direct antihelminthic function of IL-27 on γδ T cells as adoptive intratracheal transfer of rmIL-27-treated γδ T cells during primary N. brasiliensis lung infection conferred protection in mice. In summary, this report demonstrates protective functions of IL-27 to control the early lung larval stage of hookworm infection.
Collapse
Affiliation(s)
- Arjun Sharma
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jason B Noon
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Lucien P Garo
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Johannes Platten
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lee J Quinton
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joseph F Urban
- Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory and Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, U.S. Department of Agriculture, Beltsville, MD; and
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts;
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
28
|
Chen X, Liu F, Zheng B, Kang X, Wang X, Mou W, Zhang H, Jiao A, Zhao S, Gui J. Exhausted and Apoptotic BALF T Cells in Proinflammatory Airway Milieu at Acute Phase of Severe Mycoplasma Pneumoniae Pneumonia in Children. Front Immunol 2022; 12:760488. [PMID: 35111152 PMCID: PMC8801936 DOI: 10.3389/fimmu.2021.760488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022] Open
Abstract
Severe mycoplasma pneumoniae pneumonia (MPP) in children presents with serious clinical complications. Without proper and prompt intervention, it could lead to deadly consequences. Dynamics of the inflammatory airway milieu and activation status of immune cells were believed to be the hallmark of the pathogenesis and progress of the disease. In this study, by employing the T-cell sorting and mRNA microarray, we were able to define the main feature of the chemokine/cytokine expression and the unique characteristics of T cells in the bronchoalveolar lavage fluid (BALF) from severe MPP patients at acute phase. Our study for the first time delineated the molecular changes in isolated BALF T cells in severe MPP children with respect to the cytokine/chemokine expression, cell activation, exhaustion, and apoptosis. By comparing the BALF aqueous expression of cytokines/chemokines with that in sorted T cells, our data give a preliminary clue capable of finishing out the possible cell source of the proinflammatory cytokines/chemokines from the BALF mixture. Meanwhile, our data provide a distinctively pellucid expression profile particularly belonging to the isolated BALF T cells demonstrating that in the inflammatory airway, overactivated T cells were exhausted and on the verge of apoptotic progress.
Collapse
Affiliation(s)
- Xi Chen
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital affiliated to the Capital Medical University, National Center for Children's Health, Beijing, China
| | - Fang Liu
- Department of Interventional Pulmonology, Beijing Children's Hospital affiliated to the Capital Medical University, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Baoying Zheng
- Department of Pulmonology, The Children's Hospital Affiliated to the Capital Institute of Pediatrics, Beijing, China
| | - Xiaohui Kang
- Department of Pulmonology, The Children's Hospital Affiliated to the Capital Institute of Pediatrics, Beijing, China
| | - Xiaolin Wang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital affiliated to the Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital affiliated to the Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hui Zhang
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital affiliated to the Capital Medical University, National Center for Children's Health, Beijing, China
| | - Anxia Jiao
- Department of Interventional Pulmonology, Beijing Children's Hospital affiliated to the Capital Medical University, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Shunying Zhao
- Department of Respiratory Diseases, Beijing Children's Hospital affiliated to the Capital Medical University, National Clinical Research Center for Respiratory Diseases, National Center for Children's Health, Beijing, China
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital affiliated to the Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
29
|
Liu C, Hu F, Jiao G, Guo Y, Zhou P, Zhang Y, Zhang Z, Yi J, You Y, Li Z, Wang H, Zhang X. Dental pulp stem cell-derived exosomes suppress M1 macrophage polarization through the ROS-MAPK-NFκB P65 signaling pathway after spinal cord injury. J Nanobiotechnology 2022; 20:65. [PMID: 35109874 PMCID: PMC8811988 DOI: 10.1186/s12951-022-01273-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/17/2022] [Indexed: 12/31/2022] Open
Abstract
Stem cell-derived exosomes have recently been regarded as potential drugs for treating spinal cord injury (SCI) by reducing reactive oxygen species (ROS) and suppressing M1 macrophage polarization. However, the roles of ROS and exosomes in the process of M1 macrophage polarization are not known. Herein, we demonstrated that ROS can induce M1 macrophage polarization and have a concentration-dependent effect. ROS can induce M1 macrophage polarization through the MAPK-NFκB P65 signaling pathway. Dental pulp stem cell (DPSC)-derived exosomes can reduce macrophage M1 polarization through the ROS-MAPK-NFκB P65 signaling pathway in treating SCI. This study suggested that DPSC-derived exosomes might be a potential drug for treating SCI. Disruption of the cycle between ROS and M1 macrophage polarization might also be a potential effective treatment by reducing secondary damage.
Collapse
Affiliation(s)
- Chao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Fanqi Hu
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Genlong Jiao
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Yue Guo
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Pan Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
| | - Yuning Zhang
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Zhen Zhang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Jing Yi
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Yonggang You
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Zhizhong Li
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Huangpu Avenue West Road, Guangzhou, People's Republic of China.
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.
| | - Xuesong Zhang
- Department of Orthopaedics, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China.
| |
Collapse
|
30
|
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Semin Immunol 2022; 59:101629. [PMID: 35753867 DOI: 10.1016/j.smim.2022.101629] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
31
|
Rong Y, Ji C, Wang Z, Ge X, Wang J, Ye W, Tang P, Jiang D, Fan J, Yin G, Liu W, Cai W. Small extracellular vesicles encapsulating CCL2 from activated astrocytes induce microglial activation and neuronal apoptosis after traumatic spinal cord injury. J Neuroinflammation 2021; 18:196. [PMID: 34511129 PMCID: PMC8436564 DOI: 10.1186/s12974-021-02268-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023] Open
Abstract
Background Spinal cord injury (SCI) is a severe traumatic disease which causes high disability and mortality rates. The molecular pathological features after spinal cord injury mainly involve the inflammatory response, microglial and neuronal apoptosis, abnormal proliferation of astrocytes, and the formation of glial scars. However, the microenvironmental changes after spinal cord injury are complex, and the interactions between glial cells and nerve cells remain unclear. Small extracellular vesicles (sEVs) may play a key role in cell communication by transporting RNA, proteins, and bioactive lipids between cells. Few studies have examined the intercellular communication of astrocytes through sEVs after SCI. The inflammatory signal released from astrocytes is known to initiate microglial activation, but its effects on neurons after SCI remain to be further clarified. Methods Electron microscopy (TEM), nanoparticle tracking analysis (NTA), and western blotting were applied to characterize sEVs. We examined microglial activation and neuronal apoptosis mediated by astrocyte activation in an experimental model of acute spinal cord injury and in cell culture in vitro. Results Our results indicated that astrocytes activated after spinal cord injury release CCL2, act on microglia and neuronal cells through the sEV pathway, and promote neuronal apoptosis and microglial activation after binding the CCR2. Subsequently, the activated microglia release IL-1β, which acts on neuronal cells, thereby further aggravating their apoptosis. Conclusion This study elucidates that astrocytes interact with microglia and neurons through the sEV pathway after SCI, enriching the mechanism of CCL2 in neuroinflammation and spinal neurodegeneration, and providing a new theoretical basis of CCL2 as a therapeutic target for SCI. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02268-y.
Collapse
Affiliation(s)
- Yuluo Rong
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chengyue Ji
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhuanghui Wang
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xuhui Ge
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wu Ye
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Pengyu Tang
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Dongdong Jiang
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jin Fan
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wei Liu
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Weihua Cai
- Department of Orthopaedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
32
|
Yu L, Zhang Y, Xiong J, Liu J, Zha Y, Kang Q, Zhi P, Wang Q, Wang H, Zeng W, Huang Y. Activated γδ T Cells With Higher CD107a Expression and Inflammatory Potential During Early Pregnancy in Patients With Recurrent Spontaneous Abortion. Front Immunol 2021; 12:724662. [PMID: 34484234 PMCID: PMC8416064 DOI: 10.3389/fimmu.2021.724662] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
Previous studies have reported the involvement of γδ T cells in recurrent spontaneous abortion (RSA); however, both pathogenic and protective effects were suggested. To interrogate the role of γδ T cells in RSA, peripheral blood from RSA patients and healthy women with or without pregnancy were analyzed for γδ T cells by flow cytometry (n = 9–11 for each group). Moreover, the decidua from pregnant RSA patients and healthy controls (RSA-P and HC-P group, respectively) was simultaneously stained for γδ T cells by immunohistochemistry (IHC) and bulk sequenced for gene expression. Our results demonstrated that the frequencies of peripheral γδ T cells and their subpopulations in RSA patients were comparable to that in healthy subjects, but the PD1 expression on Vδ2+ cells was increased in pregnant patients. Furthermore, peripheral Vδ2+ cells in RSA-P patients demonstrated significantly increased expression of CD107a, as compared to that in pregnant healthy controls. In addition, RSA-P patients had higher proportion of IL-17A-secreting but not IL-4-secreting Vδ2+ cells compared to the control groups. In decidua, an inflammatory microenvironment was also evident in RSA-P patients, in which CCL8 expression and the infiltration of certain immune cells were higher than that in the HC-P group, as revealed by transcriptional analysis. Finally, although the presence of γδ T cells in decidua could be detected during pregnancy in both RSA patients and healthy subjects by multicolor IHC analysis, the expression of CD107a on γδ T cells was markedly higher in the RSA-P group. Collectively, our results indicated that the increased activation, cytotoxicity, and inflammatory potential of peripheral and/or local γδ T cells might be responsible for the pathogenesis of RSA. These findings could provide a better understanding of the role of γδ T cells in RSA and shed light on novel treatment strategies by targeting γδ T cells for RSA patients.
Collapse
Affiliation(s)
- Long Yu
- Department of Immunology, Medical College, Wuhan University of Science and Technology, Wuhan, China.,Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, The Second Hospital of Chaoyang City, Chaoyang, China
| | - Jinfeng Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianjun Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zha
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Kang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Zhi
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Wang
- Department of Immunology, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanjiang Zeng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|