1
|
Liu Y, Cai X, Shi B, Mo Y, Zhang J, Luo W, Yu B, Li X. Mechanisms and Therapeutic Prospects of Microglia-Astrocyte Interactions in Neuropathic Pain Following Spinal Cord Injury. Mol Neurobiol 2025; 62:4654-4676. [PMID: 39470872 DOI: 10.1007/s12035-024-04562-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
Neuropathic pain is a prevalent and debilitating condition experienced by the majority of individuals with spinal cord injury (SCI). The complex pathophysiology of neuropathic pain, involving continuous activation of microglia and astrocytes, reactive gliosis, and altered neuronal plasticity, poses significant challenges for effective treatment. This review focuses on the pivotal roles of microglia and astrocytes, the two major glial cell types in the central nervous system, in the development and maintenance of neuropathic pain after SCI. We highlight the extensive bidirectional interactions between these cells, mediated by the release of inflammatory mediators, neurotransmitters, and neurotrophic factors, which contribute to the amplification of pain signaling. Understanding the microglia-astrocyte crosstalk and its impact on neuronal function is crucial for developing novel therapeutic strategies targeting neuropathic pain. In addition, this review discusses the fundamental biology, post-injury pain roles, and therapeutic prospects of microglia and astrocytes in neuropathic pain after SCI and elucidates the specific signaling pathways involved. We also speculated that the extracellular matrix (ECM) can affect the glial cells as well. Furthermore, we also mentioned potential targeted therapies, challenges, and progress in clinical trials, as well as new biomarkers and therapeutic targets. Finally, other relevant cell interactions in neuropathic pain and the role of glial cells in other neuropathic pain conditions have been discussed. This review serves as a comprehensive resource for further investigations into the microglia-astrocyte interaction and the detailed mechanisms of neuropathic pain after SCI, with the aim of improving therapeutic efficacy.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xintong Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yajie Mo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianmin Zhang
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenting Luo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
2
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [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: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Yao L, Sai HV, Shippy T, Li B. Cellular and Transcriptional Response of Human Astrocytes to Hybrid Protein Materials. ACS APPLIED BIO MATERIALS 2024; 7:2887-2898. [PMID: 38632900 DOI: 10.1021/acsabm.3c01266] [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] [Indexed: 04/19/2024]
Abstract
Collagen is a major component of the tissue matrix, and soybean can regulate the tissue immune response. Both materials have been used to fabricate biomaterials for tissue repair. In this study, adult and fetal human astrocytes were grown in a soy protein isolate (SPI)-collagen hybrid gel or on the surface of a cross-linked SPI-collagen membrane. Hybrid materials reduced the cell proliferation rate compared to materials generated by collagen alone. However, the hybrid materials did not significantly change the cell motility compared to the control collagen material. RNA-sequencing (RNA-Seq) analysis showed downregulated genes in the cell cycle pathway, including CCNA2, CCNB1, CCNB2, CCND1, CCND2, and CDK1, which may explain lower cell proliferation in the hybrid material. This study also revealed the downregulation of genes encoding extracellular matrix (ECM) components, including HSPG2, LUM, SDC2, COL4A1, COL4A5, COL4A6, and FN1, as well as genes encoding chemokines, including CCL2, CXCL1, CXCL2, CX3CL1, CXCL3, and LIF, for adult human astrocytes grown on the hybrid membrane compared with those grown on the control collagen membrane. The study explored the cellular and transcriptional responses of human astrocytes to the hybrid material and indicated a potential beneficial function of the material in the application of neural repair.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| | - Haneesha Vishwa Sai
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| | - Teresa Shippy
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, Kansas 66506, United States
| | - Bin Li
- Department of Mechanical Engineering, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260, United States
| |
Collapse
|
4
|
Xia Y, Ding L, Zhang C, Xu Q, Shi M, Gao T, Zhou FQ, Deng DYB. Inflammatory Factor IL1α Induces Aberrant Astrocyte Proliferation in Spinal Cord Injury Through the Grin2c/Ca 2+/CaMK2b Pathway. Neurosci Bull 2024; 40:421-438. [PMID: 37864744 PMCID: PMC11003951 DOI: 10.1007/s12264-023-01128-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/11/2023] [Indexed: 10/23/2023] Open
Abstract
Spinal cord injury (SCI) is one of the most devastating traumas, and the aberrant proliferation of astrocytes usually causes neurological deficits. However, the mechanism underlying astrocyte over-proliferation after SCI is unclear. Grin2c (glutamate ionotropic receptor type 2c) plays an essential role in cell proliferation. Our bioinformatic analysis indicated that Grin2c and Ca2+ transport functions were inhibited in astrocytes after SCI. Suppression of Grin2c stimulated astrocyte proliferation by inhibiting the Ca2+/calmodulin-dependent protein kinase 2b (CaMK2b) pathway in vitro. By screening different inflammatory factors, interleukin 1α (IL1α) was further found to inhibit Grin2c/Ca2+/CaMK2b and enhance astrocyte proliferation in an oxidative damage model. Blockade of IL1α using neutralizing antibody resulted in increased Grin2c expression and the inhibition of astrocyte proliferation post-SCI. Overall, this study suggests that IL1α promotes astrocyte proliferation by suppressing the Grin2c/Ca2+/CaMK2b pathway after SCI, revealing a novel pathological mechanism of astrocyte proliferation, and may provide potential targets for SCI repair.
Collapse
Affiliation(s)
- Yu Xia
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lu Ding
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Changlin Zhang
- Department of Gynecology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
- Pelvic Floor Disorders Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qi Xu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Ming Shi
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Tianshun Gao
- Big Data Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Feng-Quan Zhou
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - David Y B Deng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
- Orthopaedic and Neurological Repair Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
5
|
Agarwal G, Roy A, Singh AA, Kumar H, Mandoli A, Srivastava A. BM-MSC-Loaded Graphene-Collagen Cryogels Ameliorate Neuroinflammation in a Rat Spinal Cord Injury Model. ACS APPLIED BIO MATERIALS 2024; 7:1478-1489. [PMID: 38354406 DOI: 10.1021/acsabm.3c00876] [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: 02/16/2024]
Abstract
A major obstacle to axonal regeneration following spinal cord injury (SCI) is neuroinflammation mediated by astrocytes and microglial cells. We previously demonstrated that graphene-based collagen hydrogels alone can decrease neuroinflammation in SCI. Their regenerative potential, however, is poorly understood and incomplete. Furthermore, stem cells have demonstrated both neuroprotective and regenerative properties in spinal cord regeneration, although there are constraints connected with the application of stem cell-based therapy. In this study, we have analyzed the regeneration capability of human bone marrow mesenchymal stem cell (BM-MSC)-loaded graphene-cross-linked collagen cryogels (Gr-Col) in a thoracic (T10-T11) hemisection model of SCI. Our study found that BM-MSC-loaded Gr-Col improves axonal regeneration, reduces neuroinflammation by decreasing astrocyte reactivity, and promotes M2 macrophage polarization. BM-MSC-loaded-Gr-Col demonstrated enhanced regenerative potential compared to Gr-Col and the injury group control. Next-generation sequencing (NGS) analysis revealed that BM-MSC-loaded-Gr-Col modulates the JAK2-STAT3 pathway, thus decreasing the reactive and scar-forming astrocyte phenotype. The decrease in neuroinflammation in the BM-MSC-loaded-Gr-Col group is attributed to the modulation of Notch/Rock and STAT5a/b and STAT6 signaling. Overall, Gene Set Enrichment Analysis suggests the promising role of BM-MSC-loaded-Gr-Col in promoting axonal regeneration after SCI by modulating molecular pathways such as the PI3/Akt pathway, focal adhesion kinase, and various inflammatory pathways.
Collapse
Affiliation(s)
- Gopal Agarwal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek A Singh
- Department of Molecular Biology, Radboud University, Postbus 9101, Nijmegen 6500 HB, The Netherlands
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Akshay Srivastava
- Department of Medical Device, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
6
|
Yoshida T, Tashiro S, Nagoshi N, Shinozaki M, Shibata T, Inoue M, Ogawa S, Shibata S, Tsuji T, Okano H, Nakamura M. Chronic Spinal Cord Injury Regeneration with Combined Therapy Comprising Neural Stem/Progenitor Cell Transplantation, Rehabilitation, and Semaphorin 3A Inhibitor. eNeuro 2024; 11:ENEURO.0378-23.2024. [PMID: 38262737 PMCID: PMC10866332 DOI: 10.1523/eneuro.0378-23.2024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024] Open
Abstract
Spinal cord injury (SCI) often results in various long-term sequelae, and chronically injured spinal cords exhibit a refractory feature, showing a limited response to cell transplantation therapies. To our knowledge, no preclinical studies have reported a treatment approach with results surpassing those of treatment comprising rehabilitation alone. In this study of rats with SCI, we propose a novel combined therapy involving a semaphorin 3A inhibitor (Sema3Ai), which enhances axonal regeneration, as the third treatment element in combination with neural stem/progenitor cell transplantation and rehabilitation. This comprehensive therapeutic strategy achieved significant improvements in host-derived neuronal and oligodendrocyte differentiation at the SCI epicenter and promoted axonal regeneration even in the chronically injured spinal cord. The elongated axons established functional electrical connections, contributing to significant enhancements in locomotor mobility when compared with animals treated with transplantation and rehabilitation. As a result, our combined transplantation, Sema3Ai, and rehabilitation treatment have the potential to serve as a critical step forward for chronic SCI patients, improving their ability to regain motor function.
Collapse
Affiliation(s)
- Takashi Yoshida
- Departments of Rehabilitation Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Syoichi Tashiro
- Departments of Rehabilitation Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Narihito Nagoshi
- Orthopaedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Munehisa Shinozaki
- Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takahiro Shibata
- Orthopaedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Mitsuhiro Inoue
- Regenerative and Cellular Medicine Kobe Center, Sumitomo Pharma, Kobe, Hyogo 650-0047, Japan
| | - Shoji Ogawa
- Formulation Research & Development Laboratories, Sumitomo Pharma, Suita, Osaka 564-0053, Japan
| | - Shinsuke Shibata
- Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata-shi, Niigata 951-8510, Japan
| | - Tetsuya Tsuji
- Departments of Rehabilitation Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Orthopaedic Surgery, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
7
|
Hendler RM, Weiss OE, Morad T, Sion G, Kirby M, Dubinsky Z, Barbora A, Minnes R, Baranes D. A Poly-D-lysine-Coated Coralline Matrix Promotes Hippocampal Neural Precursor Cells' Differentiation into GFAP-Positive Astrocytes. Polymers (Basel) 2023; 15:4054. [PMID: 37896298 PMCID: PMC10610048 DOI: 10.3390/polym15204054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
A major goal of regenerative medicine of the central nervous system is to accelerate the regeneration of nerve tissue, where astrocytes, despite their positive and negative roles, play a critical role. Thus, scaffolds capable of producing astrocytes from neural precursor cells (NPCs) are most desirable. Our study shows that NPCs are converted into reactive astrocytes upon cultivation on coralline-derived calcium carbonate coated with poly-D-lysine (PDL-CS). As shown via nuclei staining, the adhesion of neurospheres containing hundreds of hippocampal neural cells to PDL-CS resulted in disaggregation of the cell cluster as well as the radial migration of dozens of cells away from the neurosphere core. Migrating cells per neurosphere averaged 100 on PDL-CS, significantly higher than on uncoated CS (28), PDL-coated glass (65), or uncoated glass (20). After 3 days of culture on PDL-CS, cell migration plateaued and remained stable for four more days. In addition, NPCs expressing nestin underwent continuous morphological changes from round to spiky, extending and elongating their processes, resembling activated astrocytes. The extension of the process increased continuously during the maturation of the culture and doubled after 7 days compared to day 1, whereas bifurcation increased by twofold during the first 3 days before plateauing. In addition, nestin positive cells' shape, measured through the opposite circularity level correlation, decreased approximately twofold after three days, indicating spiky transformation. Moreover, nestin-positive cells co-expressing GFAP increased by 2.2 from day 1 to 7, reaching 40% of the NPC population on day 7. In this way, PDL-CS promotes NPC differentiation into reactive astrocytes, which could accelerate the repair of neural tissue.
Collapse
Affiliation(s)
- Roni Mina Hendler
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Orly Eva Weiss
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Tzachy Morad
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
| | - Guy Sion
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| | - Michael Kirby
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
- Adelson School of Medicine, Ariel University, Ariel 4070000, Israel
| | - Zvy Dubinsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Ayan Barbora
- Department of Physics, Ariel University, Ariel 4070000, Israel
| | - Refael Minnes
- Department of Physics, Ariel University, Ariel 4070000, Israel
| | - Danny Baranes
- Department of Molecular Biology, Ariel University, Ariel 4070000, Israel
- Adelson School of Medicine, Ariel University, Ariel 4070000, Israel
| |
Collapse
|
8
|
Chu PH, Chen SC, Chen HY, Wu CB, Huang WT, Chiang HY. Astrocyte-associated fibronectin promotes the proinflammatory phenotype of astrocytes through β1 integrin activation. Mol Cell Neurosci 2023; 125:103848. [PMID: 36948232 DOI: 10.1016/j.mcn.2023.103848] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023] Open
Abstract
Astrocytes are key players in neuroinflammation. In response to central nervous system (CNS) injury or disease, astrocytes undergo reactive astrogliosis, which is characterized by increased proliferation, migration, and glial fibrillary acidic protein (GFAP) expression. Activation of the transcription factor nuclear factor-κB (NF-κB) and upregulation of downstream proinflammatory mediators in reactive astrocytes induce a proinflammatory phenotype in astrocytes, thereby exacerbating neuroinflammation by establishing an inflammatory loop. In this study, we hypothesized that excessive fibronectin (FN) derived from reactive astrocytes would induce this proinflammatory phenotype in astrocytes in an autocrine manner. We exogenously treated astrocytes with monomer FN, which can be incorporated into the extracellular matrix (ECM), to mimic plasma FN extravasated through a compromised blood-brain barrier in neuroinflammation. We also induced de novo synthesis and accumulation of astrocyte-derived FN through tumor necrosis factor-α (TNF-α) stimulation. The excessive FN deposition resulting from both treatments initiated reactive astrogliosis and triggered NF-κB signaling in the cultured astrocytes. In addition, inhibition of FN accumulation in the ECM by the FN inhibitor pUR4 strongly attenuated the FN- and TNF-α-induced GFAP expression, NF-κB activation, and proinflammatory mediator production of astrocytes by interrupting FN-β1 integrin coupling and thus the inflammatory loop. In an in vivo experiment, intrathecal injection of pUR4 considerably ameliorated FN deposition, GFAP expression, and NF-κB activation in inflamed spinal cord, suggesting the therapeutic potential of pUR4 for attenuating neuroinflammation and promoting neuronal function restoration.
Collapse
Affiliation(s)
- Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Shao-Chi Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Yung Chen
- Department of Occupational Therapy, Graduate Institute of Behavioral Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Bei Wu
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Ting Huang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
9
|
Jin C, Wu Y, Zhang H, Xu B, Liu W, Ji C, Li P, Chen Z, Chen B, Li J, Wu X, Jiang P, Hu Y, Xiao Z, Zhao Y, Dai J. Spinal cord tissue engineering using human primary neural progenitor cells and astrocytes. Bioeng Transl Med 2023; 8:e10448. [PMID: 36925694 PMCID: PMC10013752 DOI: 10.1002/btm2.10448] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 11/11/2022] Open
Abstract
Neural progenitor cell (NPC) transplantation is a promising approach for repairing spinal cord injury (SCI). However, cell survival, maturation and integration after transplantation are still major challenges. Here, we produced a novel centimeter-scale human spinal cord neural tissue (hscNT) construct with human spinal cord neural progenitor cells (hscNPCs) and human spinal cord astrocytes (hscAS) on a linearly ordered collagen scaffold (LOCS). The hscAS promoted hscNPC adhesion, survival and neurite outgrowth on the LOCS, to form a linearly ordered spinal cord-like structure consisting of mature neurons and glia cells. When transplanted into rats with SCI, the hscNT created a favorable microenvironment by inhibiting inflammation and glial scar formation, and promoted neural and vascular regeneration. Notably, the hscNT promoted neural circuit reconstruction and motor functional recovery. Engineered human spinal cord implants containing astrocytes and neurons assembled on axon guidance scaffolds may therefore have potential in the treatment of SCI.
Collapse
Affiliation(s)
- Chen Jin
- University of the Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Yayu Wu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Haipeng Zhang
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Bai Xu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Wenbin Liu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Chunnan Ji
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Panpan Li
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Zhenni Chen
- University of the Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Bing Chen
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Jiayin Li
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Peipei Jiang
- Department of Obstetrics and GynecologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yali Hu
- Department of Obstetrics and GynecologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| | - Jianwu Dai
- University of the Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Molecular Developmental BiologyInstitute of Genetics and Developmental Biology, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
10
|
Tamaru T, Kobayakawa K, Saiwai H, Konno D, Kijima K, Yoshizaki S, Hata K, Iura H, Ono G, Haruta Y, Kitade K, Iida KI, Kawaguchi KI, Matsumoto Y, Kubota K, Maeda T, Okada S, Nakashima Y. Glial scar survives until the chronic phase by recruiting scar-forming astrocytes after spinal cord injury. Exp Neurol 2023; 359:114264. [PMID: 36336030 DOI: 10.1016/j.expneurol.2022.114264] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Spinal cord injury (SCI) causes reactive astrogliosis, the sequential phenotypic change of astrocytes in which naïve astrocytes (NAs) transform into reactive astrocytes (RAs) and subsequently become scar-forming astrocytes (SAs), resulting in glial scar formation around the lesion site and thereby limiting axonal regeneration and motor/sensory functional recovery. Inhibiting the transformation of RAs into SAs in the acute phase attenuates the reactive astrogliosis and promotes regeneration. However, whether or not SAs once formed can revert to RAs or SAs is unclear. We performed selective isolation of astrocytes from glial scars at different time points for a gene expression analysis and found that the expression of Sox9, an important transcriptional factor for glial cell differentiation, was significantly increased in chronic phase astrocytes (CAs) compared to SAs in the sub-acute phase. Furthermore, CAs showed a significantly lower expression of chondroitin sulfate proteoglycan (CSPG)-related genes than SAs. These results indicated that SAs changed their phenotypes according to the surrounding environment of the injured spinal cord over time. Even though the integrin-N-cadherin pathway is critical for glial scar formation, collagen-I-grown scar-forming astrocytes (Col-I-SAs) did not change their phenotype after depleting the effect of integrin or N-cadherin. In addition, we found that Col-I-SAs transplanted into a naïve spinal cord formed glial scar again by maintaining a high expression of genes involved in the integrin-N-cadherin pathway and a low expression of CSPG-related genes. Interestingly, the transplanted Col-I-SAs changed NAs into SAs, and anti-β1-integrin antibody blocked the recruitment of SAs while reducing the volume of glial scar in the chronic phase. Our findings indicate that while the characteristics of glial scars change over time after SCI, SAs have a cell-autonomous function to form and maintain a glial scar, highlighting the basic mechanism underlying the persistence of glial scars after central nervous system injury until the chronic phase, which may be a therapeutic target.
Collapse
Affiliation(s)
- Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering Kindai University, Higashiosaka, Japan
| | - Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Orthopedic Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Kazuhiro Hata
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei-Ichiro Iida
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-Ichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Kubota
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Takeshi Maeda
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Osaka University, Suita, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
11
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
12
|
Bugiani M, Plug BC, Man JHK, Breur M, van der Knaap MS. Heterogeneity of white matter astrocytes in the human brain. Acta Neuropathol 2022; 143:159-177. [PMID: 34878591 DOI: 10.1007/s00401-021-02391-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Astrocytes regulate central nervous system development, maintain its homeostasis and orchestrate repair upon injury. Emerging evidence support functional specialization of astroglia, both between and within brain regions. Different subtypes of gray matter astrocytes have been identified, yet molecular and functional diversity of white matter astrocytes remains largely unexplored. Nonetheless, their important and diverse roles in maintaining white matter integrity and function are well recognized. Compelling evidence indicate that impairment of normal astrocytic function and their response to injury contribute to a wide variety of diseases, including white matter disorders. In this review, we highlight our current understanding of astrocyte heterogeneity in the white matter of the mammalian brain and how an interplay between developmental origins and local environmental cues contribute to astroglial diversification. In addition, we discuss whether, and if so, how, heterogeneous astrocytes could contribute to white matter function in health and disease and focus on the sparse human research data available. We highlight four leukodystrophies primarily due to astrocytic dysfunction, the so-called astrocytopathies. Insight into the role of astroglial heterogeneity in both healthy and diseased white matter may provide new avenues for therapies aimed at promoting repair and restoring normal white matter function.
Collapse
|
13
|
Ikeshima-Kataoka H, Sugimoto C, Tsubokawa T. Integrin Signaling in the Central Nervous System in Animals and Human Brain Diseases. Int J Mol Sci 2022; 23:ijms23031435. [PMID: 35163359 PMCID: PMC8836133 DOI: 10.3390/ijms23031435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
The integrin family is involved in various biological functions, including cell proliferation, differentiation and migration, and also in the pathogenesis of disease. Integrins are multifunctional receptors that exist as heterodimers composed of α and β subunits and bind to various ligands, including extracellular matrix (ECM) proteins; they are found in many animals, not only vertebrates (e.g., mouse, rat, and teleost fish), but also invertebrates (e.g., planarian flatworm, fruit fly, nematodes, and cephalopods), which are used for research on genetics and social behaviors or as models for human diseases. In the present paper, we describe the results of a phylogenetic tree analysis of the integrin family among these species. We summarize integrin signaling in teleost fish, which serves as an excellent model for the study of regenerative systems and possesses the ability for replacing missing tissues, especially in the central nervous system, which has not been demonstrated in mammals. In addition, functions of astrocytes and reactive astrocytes, which contain neuroprotective subpopulations that act in concert with the ECM proteins tenascin C and osteopontin via integrin are also reviewed. Drug development research using integrin as a therapeutic target could result in breakthroughs for the treatment of neurodegenerative diseases and brain injury in mammals.
Collapse
Affiliation(s)
- Hiroko Ikeshima-Kataoka
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
- Faculty of Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
- Correspondence:
| | - Chikatoshi Sugimoto
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
| | - Tatsuya Tsubokawa
- Department of Biology, Keio University, 4-1-1, Hiyoshi, Kohoku-ku, Yokohama-shi 223-8521, Japan; (C.S.); (T.T.)
| |
Collapse
|
14
|
Wu J, Zhu ZY, Fan ZW, Chen Y, Yang RY, Li Y. Downregulation of EphB2 by RNA interference attenuates glial/fibrotic scar formation and promotes axon growth. Neural Regen Res 2022; 17:362-369. [PMID: 34269211 PMCID: PMC8463997 DOI: 10.4103/1673-5374.317988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The rapid formation of a glial/fibrotic scar is one of the main factors hampering axon growth after spinal cord injury. The bidirectional EphB2/ephrin-B2 signaling of the fibroblast-astrocyte contact-dependent interaction is a trigger for glial/fibrotic scar formation. In the present study, a new in vitro model was produced by coculture of fibroblasts and astrocytes wounded by scratching to mimic glial/fibrotic scar-like structures using an improved slide system. After treatment with RNAi to downregulate EphB2, changes in glial/fibrotic scar formation and the growth of VSC4.1 motoneuron axons were examined. Following RNAi treatment, fibroblasts and astrocytes dispersed without forming a glial/fibrotic scar-like structure. Furthermore, the expression levels of neurocan, NG2 and collagen I in the coculture were reduced, and the growth of VSC4.1 motoneuron axons was enhanced. These findings suggest that suppression of EphB2 expression by RNAi attenuates the formation of a glial/fibrotic scar and promotes axon growth. This study was approved by the Laboratory Animal Ethics Committee of Jiangsu Province, China (approval No. 2019-0506-002) on May 6, 2019.
Collapse
Affiliation(s)
- Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhen-Yu Zhu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhi-Wei Fan
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ri-Yun Yang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
15
|
Yao S, Wang L, Chen Q, Lu T, Pu X, Luo C. The effect of mild hypothermia plus rutin on the treatment of spinal cord injury and inflammatory factors by repressing TGF-β/smad pathway. Acta Cir Bras 2021; 36:e360307. [PMID: 33978063 PMCID: PMC8112105 DOI: 10.1590/acb360307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Purpose To probe the mechanism of mild hypothermia combined with rutin in the
treatment of spinal cord injury (SCI). Methods Thirty rats were randomized into the following groups: control, sham, model,
mild hypothermia (MH), and mild hypothermia plus rutin (MH+Rutin). We used
modified Allen’s method to injure the spinal cord (T10) in rats, and then
treated it with MH or/and rutin immediately. BBB scores were performed on
all rats. We used HE staining for observing the injured spinal cord tissue;
ELISA for assaying TNF-α, IL-1β, IL-8, Myeloperoxidase (MPO), and
Malondialdehyde (MDA) contents; Dihydroethidium (DHE) for measuring the
reactive oxygen species (ROS) content; flow cytometry for detecting
apoptosis; and both RT-qPCR and Western blot for determining the expression
levels of TGF-β/Smad pathway related proteins (TGF-β, Smad2, and Smad3). Results In comparison with model group, the BBB score of MH increased to a certain
extent and MH+Rutin group increased more than MH group (p < 0.05). After
treatment with MH and MH+Rutin, the inflammatory infiltration diminished. MH
and MH+Rutin tellingly dwindled TNF-β, MDA and ROS contents (p < 0.01),
and minified spinal cord cell apoptosis. MH and MH+Rutin could patently
diminished TGF-β1, Smad2, and Smad3 expression (p < 0.01). Conclusions MH+Rutin can suppress the activation of TGF-β/Smad pathway, hence repressing
the cellular inflammatory response after SCI.
Collapse
|
16
|
Parra-Villamar D, Blancas-Espinoza L, Garcia-Vences E, Herrera-García J, Flores-Romero A, Toscano-Zapien A, Villa JV, Barrera-Roxana R, Karla SZ, Ibarra A, Silva-García R. Neuroprotective effect of immunomodulatory peptides in rats with traumatic spinal cord injury. Neural Regen Res 2021; 16:1273-1280. [PMID: 33318405 PMCID: PMC8284257 DOI: 10.4103/1673-5374.301485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Several therapies have shown obvious effects on structural conservation contributing to motor functional recovery after spinal cord injury (SCI). Nevertheless, neither strategy has achieved a convincing effect. We purposed a combined therapy of immunomodulatory peptides that individually have shown significant effects on motor functional recovery in rats with SCI. The objective of this study was to investigate the effects of the combined therapy of monocyte locomotion inhibitor factor (MLIF), A91 peptide, and glutathione monoethyl ester (GSH-MEE) on chronic-stage spinal cord injury. Female Sprague-Dawley rats underwent a laminectomy of the T9 vertebra and a moderate contusion. Six groups were included: sham, PBS, MLIF + A91, MLIF + GSH-MEE, A91 + GSH-MEE, and MLIF + A91 + GSH-MEE. Two months after injury, motor functional recovery was evaluated using the open field test. Parenchyma and white matter preservation was evaluated using hematoxylin & eosin staining and Luxol Fast Blue staining, respectively. The number of motoneurons in the ventral horn and the number of axonal fibers were determined using hematoxylin & eosin staining and immunohistochemistry, respectively. Collagen deposition was evaluated using Masson's trichrome staining. The combined therapy of MLIF, A91, and GSH-MEE greatly contributed to motor functional recovery and preservation of the medullary parenchyma, white matter, motoneurons, and axonal fibres, and reduced the deposition of collagen in the lesioned area. The combined therapy of MLIF, A91, and GSH-MEE preserved spinal cord tissue integrity and promoted motor functional recovery of rats after SCI. This study was approved by the National Commission for Scientific Research on Bioethics and Biosafety of the Instituto Mexicano del Seguro Social under registration number R-2015-785-116 (approval date November 30, 2015) and R-2017-3603-33 (approval date June 5, 2017).
Collapse
Affiliation(s)
- Dulce Parra-Villamar
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| | - Liliana Blancas-Espinoza
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| | - Elisa Garcia-Vences
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Campus Norte, Huixquilucan, Edo de México; Centro de Investigación del Proyecto Camina A.C, Ciudad de México, México
| | - Juan Herrera-García
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| | - Adrian Flores-Romero
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Campus Norte, Huixquilucan, Edo de México; Centro de Investigación del Proyecto Camina A.C, Ciudad de México, México
| | - Alberto Toscano-Zapien
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| | - Jonathan Vilchis Villa
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| | - Rodríguez Barrera-Roxana
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Campus Norte, Huixquilucan, Edo de México; Centro de Investigación del Proyecto Camina A.C, Ciudad de México, México
| | - Soria Zavala Karla
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México; Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Campus Norte, Huixquilucan, Edo de México, México
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México, Campus Norte, Huixquilucan, Edo de México; Centro de Investigación del Proyecto Camina A.C, Ciudad de México, México
| | - Raúl Silva-García
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, Centro Médico Nacional Siglo XXI; Instituto Mexicano del Seguro Social; Ciudad de México, México
| |
Collapse
|
17
|
Kumar M, Adeluyi A, Anderson EL, Turner JR. Glial cells as therapeutic targets for smoking cessation. Neuropharmacology 2020; 175:108157. [PMID: 32461156 PMCID: PMC7791555 DOI: 10.1016/j.neuropharm.2020.108157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/17/2022]
Abstract
Smoking remains the leading cause of morbidity and mortality in the United States, with less than 5% of smokers attempting to quit succeeding. This low smoking cessation success rate is thought to be due to the long-term adaptations and alterations in synaptic plasticity that occur following chronic nicotine exposure and withdrawal. Glial cells have recently emerged as active players in the development of dependence phenotypes due to their roles in modulating neuronal functions and synaptic plasticity. Fundamental studies have demonstrated that microglia and astrocytes are crucial for synapse formation and elimination in the developing brain, likely contributing to why glial dysfunction is implicated in numerous neurological and psychiatric disorders. Recently, there is increasing evidence for the involvement of glial cells in drug dependence and its associated behavioral manifestations. This review summarizes the newly evaluated role of microglia and astrocytes as molecular drivers of nicotine dependence and withdrawal phenotypes. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA.
| | - Adewale Adeluyi
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA; Ann Romney Center for Neurologic Disease, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Erin L Anderson
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, USA.
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, KY, USA.
| |
Collapse
|
18
|
Daverey A, Agrawal SK. Curcumin Protects against White Matter Injury through NF-κB and Nrf2 Cross Talk. J Neurotrauma 2020; 37:1255-1265. [PMID: 31914858 DOI: 10.1089/neu.2019.6749] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inflammation and oxidative stress play a central role in the pathogenesis of white matter injury (WMI). Curcumin (Cur), a polyphenolic compound, exhibits anti-inflammatory and anti-oxidant effects on several conditions. The objective of this study was to investigate neuroprotective effects of Cur on WMI and explore its underlying mechanisms of action. Sprague-Dawley rats were subjected to the removal of white matter from the dorsal column of the spinal cord. Dorsal columns were randomly divided into three groups: Sham (Ringer's solution bubbled with 95% O2 and 5% CO2), hypoxia (Hyp; Ringer's solution bubbled with 95% N2 and 5% CO2 for 1 h), and Cur-treated (Hyp+Cur; Ringer's solution bubbled with 95% N2 and 5% CO2 for 1 h in the presence of 50 μM Cur). For NF-κB inhibition experiments, dorsal columns were incubated with 50 μM BAY 11-7082 (BAY) for 30 min in 95% O2 and 5% CO2 prior to 1-h incubation with 50 μM Cur in 95% N2 and 5% CO2. Our data show that Cur inhibited hypoxia-induced HIF1-α expression and tissue damage by demonstrating the improved morphology of astrocytes and remarkable reduction in vacuolation. Cur also inhibited the hypoxia-induced upregulation of glial fibrillary acidic protein (GFAP) and neurofilament-H (NF-H) after hypoxia and downregulated the expression of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 1 (IL-1). Terminal dexynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-assay analysis showed that Cur effectively attenuated apoptosis in white matter. In addition, we demonstrated that Cur exerted its neuroprotective effect through cross talk between nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways. In conclusion, our results indicate that treatment with Cur inhibited the hypoxia, inflammation and apoptosis associated with WMI. Further, the Nrf-2 pathway inhibits NF-κB activation by preventing IkB degradation and increasing HO-1 expression, which in turn reduces reactive oxygen species (ROS) and as a result NF-κB activation is suppressed. Similarly, NF-κB-mediated transcription reduces Nrf2 activation by reducing anti-oxidant response element (ARE) gene and free CREB binding protein by competing with Nrf2 for CBP thus inhibiting the Nrf-2 activation.
Collapse
Affiliation(s)
- Amita Daverey
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sandeep K Agrawal
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
19
|
Vismara I, Papa S, Veneruso V, Mauri E, Mariani A, De Paola M, Affatato R, Rossetti A, Sponchioni M, Moscatelli D, Sacchetti A, Rossi F, Forloni G, Veglianese P. Selective Modulation of A1 Astrocytes by Drug-Loaded Nano-Structured Gel in Spinal Cord Injury. ACS NANO 2020; 14:360-371. [PMID: 31887011 DOI: 10.1021/acsnano.9b05579] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astrogliosis has a very dynamic response during the progression of spinal cord injury, with beneficial or detrimental effects on recovery. It is therefore important to develop strategies to target activated astrocytes and their harmful molecular mechanisms so as to promote a protective environment to counteract the progression of the secondary injury. The challenge is to formulate an effective therapy with maximum protective effects, but reduced side effects. In this study, a functionalized nanogel-based nanovector was selectively internalized in activated mouse or human astrocytes. Rolipram, an anti-inflammatory drug, when administered by these nanovectors limited the inflammatory response in A1 astrocytes, reducing iNOS and Lcn2, which in turn reverses the toxic effect of proinflammatory astrocytes on motor neurons in vitro, showing advantages over conventionally administered anti-inflammatory therapy. When tested acutely in a spinal cord injury mouse model, it improved motor performance, but only in the early stage after injury, reducing the astrocytosis and preserving neuronal cells.
Collapse
Affiliation(s)
- Irma Vismara
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Simonetta Papa
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Valeria Veneruso
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Emanuele Mauri
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Alessandro Mariani
- Department of Environmental Health Sciences , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Massimiliano De Paola
- Department of Environmental Health Sciences , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Roberta Affatato
- Department of Oncology , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Arianna Rossetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Mattia Sponchioni
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Davide Moscatelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Alessandro Sacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Gianluigi Forloni
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Pietro Veglianese
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| |
Collapse
|
20
|
Lee EJ, Han JC, Park DY, Kee C. A neuroglia-based interpretation of glaucomatous neuroretinal rim thinning in the optic nerve head. Prog Retin Eye Res 2020; 77:100840. [PMID: 31982595 DOI: 10.1016/j.preteyeres.2020.100840] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/02/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Neuroretinal rim thinning (NRR) is a characteristic glaucomatous optic disc change. However, the precise mechanism of the rim thinning has not been completely elucidated. This review focuses on the structural role of the glioarchitecture in the formation of the glaucomatous NRR thinning. The NRR is a glia-framed structure, with honeycomb geometry and mechanically reinforced astrocyte processes along the transverse plane. When neural damage selectively involves the neuron and spares the glia, the gross structure of the tissue is preserved. The disorganization and loss of the glioarchitecture are the two hallmarks of optic nerve head (ONH) remodeling in glaucoma that leads to the thinning of NRR tissue upon axonal loss. This is in contrast to most non-glaucomatous optic neuropathies with optic disc pallor where hypertrophy of the glioarchitecture is associated with the seemingly absent optic disc cupping. Arteritic anterior ischemic optic neuropathy is an exception where pan-necrosis of ONH tissue leads to NRR thinning. Milder ischemia indicates selective neuronal loss that spares glia in non-arteritic anterior ischemic optic neuropathy. The biological reason is the heterogeneous glial response determined by the site, type, and severity of the injury. The neuroglial interpretation explains how the cellular changes underlie the clinical findings. Updated understandings on glial responses illustrate the mechanical, microenvironmental, and microglial modulation of activated astrocytes in glaucoma. Findings relevant to the possible mechanism of the astrocyte death in advanced glaucoma are also emerging. Ultimately, a better understanding of glaucomatous glial response may lead to glia-targeting neuroprotection in the future.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Jong Chul Han
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Do Young Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Changwon Kee
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
21
|
Hlavac N, VandeVord PJ. Astrocyte Mechano-Activation by High-Rate Overpressure Involves Alterations in Structural and Junctional Proteins. Front Neurol 2019; 10:99. [PMID: 30853931 PMCID: PMC6395392 DOI: 10.3389/fneur.2019.00099] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/24/2019] [Indexed: 11/13/2022] Open
Abstract
Primary blast neurotrauma represents a unique injury paradigm characterized by high-rate overpressure effects on brain tissue. One major hallmark of blast neurotrauma is glial reactivity, notably prolonged astrocyte activation. This cellular response has been mainly defined in primary blast neurotrauma by increased intermediate filament expression. Because the intermediate filament networks physically interface with transmembrane proteins for junctional support, it was hypothesized that cell junction regulation is altered in the reactive phenotype as well. This would have implications for downstream transcriptional regulation via signal transduction pathways like nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Therefore, a custom high-rate overpressure simulator was built for in vitro testing using mechanical conditions based on intracranial pressure measurements in a rat model of blast neurotrauma. Primary rat astrocytes were exposed to isolated high-rate mechanical stimulation to study cell junction dynamics in relation to their mechano-activation. First, a time course for "classical" features of reactivity was devised by evaluation of glial fibrillary acidic protein (GFAP) and proliferating cell nuclear antigen (PCNA) expression. This was followed by gene and protein expression for both gap junction (connexins) and anchoring junction proteins (integrins and cadherins). Signal transduction analysis was carried out by nuclear localization of two molecules, NF-κB p65 and mitogen-activated protein kinase (MAPK) p38. Results indicated significant increases in connexin-43 expression and PCNA first at 24 h post-overpressure (p < 0.05), followed by structural reactivity (via increased GFAP, p < 0.05) corresponding to increased anchoring junction dynamics at 48 h post-overpressure (p < 0.05). Moreover, increased phosphorylation of focal adhesion kinase (FAK) was observed in addition to increased nuclear localization of both p65 and p38 (p < 0.05) during the period of structural reactivity. To evaluate the transcriptional activity of p65 in the nucleus, electrophoretic mobility shift assay was conducted for a binding site on the promoter region for intracellular adhesion molecule-1 (ICAM-1), an antagonist of tight junctions. A significant increase in the interaction of nuclear proteins with the NF-κB site on the ICAM-1 corresponded to increased gene and protein expression of ICAM-1 (p < 0.05). Altogether, these results indicate multiple targets and corresponding signaling pathways which involve cell junction dynamics in the mechano-activation of astrocytes following high-rate overpressure.
Collapse
Affiliation(s)
- Nora Hlavac
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute, Blacksburg, VA, United States
| | - Pamela J VandeVord
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute, Blacksburg, VA, United States.,Department of Research, Salem Veterans Affairs Medical Center, Salem, VA, United States
| |
Collapse
|
22
|
Patel R, Muir M, Cvetkovic C, Krencik R. Concepts toward directing human astroplasticity to promote neuroregeneration. Dev Dyn 2018; 248:21-33. [DOI: 10.1002/dvdy.24655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
| | | | - Caroline Cvetkovic
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| |
Collapse
|
23
|
Janowska J, Gargas J, Ziemka-Nalecz M, Zalewska T, Buzanska L, Sypecka J. Directed glial differentiation and transdifferentiation for neural tissue regeneration. Exp Neurol 2018; 319:112813. [PMID: 30171864 DOI: 10.1016/j.expneurol.2018.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Glial cells which are indispensable for the central nervous system development and functioning, are proven to be vulnerable to a harmful influence of pathological cues and tissue misbalance. However, they are also highly sensitive to both in vitro and in vivo modulation of their commitment, differentiation, activity and even the fate-switch by different types of bioactive molecules. Since glial cells (comprising macroglia and microglia) are an abundant and heterogeneous population of neural cells, which are almost uniformly distributed in the brain and the spinal cord parenchyma, they all create a natural endogenous reservoir of cells for potential neurogenerative processes required to be initiated in response to pathophysiological cues present in the local tissue microenvironment. The past decade of intensive investigation on a spontaneous and enforced conversion of glial fate into either alternative glial (for instance from oligodendrocytes to astrocytes) or neuronal phenotypes, has considerably extended our appreciation of glial involvement in restoring the nervous tissue cytoarchitecture and its proper functions. The most effective modulators of reprogramming processes have been identified and tested in a series of pre-clinical experiments. A list of bioactive compounds which are potent in guiding in vivo cell fate conversion and driving cell differentiation includes a selection of transcription factors, microRNAs, small molecules, exosomes, morphogens and trophic factors, which are helpful in boosting the enforced neuro-or gliogenesis and promoting the subsequent cell maturation into desired phenotypes. Herein, an issue of their utility for a directed glial differentiation and transdifferentiation is discussed in the context of elaborating future therapeutic options aimed at restoring the diseased nervous tissue.
Collapse
Affiliation(s)
- Justyna Janowska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Justyna Gargas
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Teresa Zalewska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Leonora Buzanska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Stem Cell Bioengineering Unit, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Joanna Sypecka
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland.
| |
Collapse
|
24
|
Lu WC, Zhou YX, Qiao P, Zheng J, Wu Q, Shen Q. The protocadherin alpha cluster is required for axon extension and myelination in the developing central nervous system. Neural Regen Res 2018; 13:427-433. [PMID: 29623926 PMCID: PMC5900504 DOI: 10.4103/1673-5374.228724] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In adult mammals, axon regeneration after central nervous system injury is very poor, resulting in persistent functional loss. Enhancing the ability of axonal outgrowth may be a potential treatment strategy because mature neurons of the adult central nervous system may retain the intrinsic ability to regrow axons after injury. The protocadherin (Pcdh) clusters are thought to function in neuronal morphogenesis and in the assembly of neural circuitry in the brain. We cultured primary hippocampal neurons from E17.5 Pcdhα deletion (del-α) mouse embryos. After culture for 1 day, axon length was obviously shorter in del-α neurons compared with wild-type neurons. RNA sequencing of hippocampal E17.5 RNA showed that expression levels of BDNF, Fmod, Nrp2, OGN, and Sema3d, which are associated with axon extension, were significantly down-regulated in the absence of the Pcdhα gene cluster. Using transmission electron microscopy, the ratio of myelinated nerve fibers in the axons of del-α hippocampal neurons was significantly decreased; myelin sheaths of P21 Pcdhα-del mice showed lamellar disorder, discrete appearance, and vacuoles. These results indicate that the Pcdhα cluster can promote the growth and myelination of axons in the neurodevelopmental stage.
Collapse
Affiliation(s)
- Wen-Cheng Lu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Xiao Zhou
- Center for Comparative Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Institute of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Qiao
- Department of Orthopedics, People's Hospital of Zhangqiu, Zhangqiu, Shandong Province, China
| | - Jin Zheng
- Center for Comparative Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Institute of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Wu
- Center for Comparative Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Institute of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Shen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|